AU2003223093A1 - Methods and compositions for treatment of central and peripheral nervous system disorders and novel compounds useful therefor - Google Patents

Methods and compositions for treatment of central and peripheral nervous system disorders and novel compounds useful therefor Download PDF

Info

Publication number
AU2003223093A1
AU2003223093A1 AU2003223093A AU2003223093A AU2003223093A1 AU 2003223093 A1 AU2003223093 A1 AU 2003223093A1 AU 2003223093 A AU2003223093 A AU 2003223093A AU 2003223093 A AU2003223093 A AU 2003223093A AU 2003223093 A1 AU2003223093 A1 AU 2003223093A1
Authority
AU
Australia
Prior art keywords
pharmaceutically acceptable
compound
spiro
thia
pharmaceutical composition
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2003223093A
Other versions
AU2003223093B2 (en
Inventor
Nira Bar-Ner
Abraham Fisher
Yishai Karton
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Israel Institute for Biological Research
Original Assignee
Israel Institute for Biological Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Israel Institute for Biological Research filed Critical Israel Institute for Biological Research
Publication of AU2003223093A1 publication Critical patent/AU2003223093A1/en
Application granted granted Critical
Publication of AU2003223093B2 publication Critical patent/AU2003223093B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/02Drugs for genital or sexual disorders; Contraceptives for disorders of the vagina
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/16Emollients or protectives, e.g. against radiation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/04Artificial tears; Irrigation solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Description

WO 03/092580 PCT/IL03/00357 1 METHODS AND COMPOSITIONS FOR TREATMENT OF CENTRAL AND PERIPHERAL NERVOUS SYSTEM DISORDERS AND NOVEL COMPOUNDS USEFUL THEREFOR Field of the Invention The invention relates to methods for treating various central and peripheral nervous system disorders. 5 Background of the invention The following documents, the contents of which are incorporated herein by reference, are believed to be relevant: Fisher and Barak. Drug News & Perspectives 7: 453-64, 1994; Review: Fisher. Jap J Pharmacol 10 84: 101-112, 2000; Wolozin et al. Arch Neurol. 57:1439-43, 2000; Sparks et al. Neurosci Lett 187:142-144, 1995; Refolo et al. Soc Neurosci Abst, San Diego, CA, USA, 2001; Refolo et al. Neurobiol Dis 8: 890-899, 2001; Review: Cedazo-Minguez and Cowburn. J Cell Mol Med 5:254-266, 2001; Bales et al. PNAS 96: 15233, 1999; Buttini et al. Neurosci 97: 207, 2000; Hartmann et al, Exp Neurol 170: 326, 2001; Mudher and Lovestone. Trends Neurosci 25:22-6, 2002; Mudher et al. J 15 Neurosci 21:4987-95, 2001; Zhang et al. Nature 395: 698-702, 1998; De Ferrari et al. Brain Res Brain Res Rev 33:1-12, 2000; Garrido et al. FASEB J 16:1982-4, 2002; Eldar-Finkelman. Trends Molec Med. 8:126-32, 2002; Bhat et al. Neurosignals 11:251-61, 2002; Gentleman et al. NeuroReport 8: 1519-1522, 1997; Roberts et al. J Neurol Neurosurg Psychiat, 57: 419-425, 1994; Havlik et al. Neurobiol Aging S140, 587, 1998; Mayeux et al. Neurol, 45: 556-557, 1995; Nicoll et al. Ann N Y 20 Acad Sci, 777: 271-275, 1996; Capruso and Levin. Neurol ClinlO: 879-893, 1992; Dixon et. al. Behav Brain Res 70:125-131, 1995; Pike and Hamm. Exptl Neurol, 147: 55-65, 1997; Pike and Hamm. Pharmacol Biochem Behav, 57: 785-791, 1997; Pike and Hamm. J Neurotrauma, 14: 897-905, 1997; U.S. Patents Nos. 4,855,290, 4,981,858, 4,900,830, 4,876,260, 5,053,412, 5,407,938, 6,277,874, 6,274,603; 25 Irwin, S. PSYCHOPHARM 13:222-257, 1968; Beach et al, Neurosci Lett 283: 9-12, 2000; Beach et al., Brain Res 905: 220-223, 2001; Pfeifer et al., Science 2002 298:1379; Nicoll et al, Nature Medicine, March 2003, Sparks et al. Neurosci Lett 1995; 187:142-144; Refolo et al Neurobiol Dis 8: 890-899, 2001 ; Refolo et al, Soc Neurosci Abst 2001, San Diego, CA, USA; Cedazo-Minguez et al, Neurosci, 105: 651-661, 2001; Sparks et al. Neurosci Lett 1995; 187:142-144; Dean et al, Mol Psychiatry 1996; 30 1:54-8; Dean et al, Mol Psychiatry 2002; 7: 1083-91; Raedler et al, Am J. Psychiatry 160: 118, 2003; Borda et al J Immunol 2002; 168:3667-74; Felder et al, Life Sci 2001 8:2605-13; Bymaster et al, Current Drug Targets- CNS & Neurological Disorders 2002; 1:147-164; Sullivan et al, Br J Psychiatry 2000; 177:177-8; Gould and Manji. Neuroscientist 2002; 8:497-511; Cotter et al, NeuroReport 9: 1379, 1998; Casanova MF et al Acta Neuropathol (Berl) 2002 103: 313-20; Auld et al Prog Neurobiol 2002, 35 68:209-45; Poeggeler et al, Brain Res 815: 382-388, 1999; Chyan et al. J Biolog Chem 274: 21937-21942, 1999; Bons et al., Alzheimer's Res (1995) 1:83-87; Mazzoti et al (Proceedings of the Chiral Europe 96 Symp, Spring Innovations, Stockport UK, p 103, 1996; Krise et al, J Med. Chem. 42: 3094-3100 (1999); WO 03/092580 PCT/IL03/00357 2 Fassbeder et al, PNAS,98: 5856, 2001; Sparks et al. Exp Neurol 1994; 126:88-94; Sparks Nutr Metab Cardiovase Dis 1997: 7:255-266; Beach et al, Neurosci Lett 283: 9-12, 2000; Beach et al Brain Res. 905: 220-223, 2001; Klausner, Biotechnol 5:779-786, 1987; Lipman et al, Cytotechnol 8:129-176, 1992; Rappoport and Ferreira J. Neurochem. 74:125-133 (2000); Ekinci et al J. Biol. Chem. 274: 5 30322-30327 (1999); Sadot et al J.Neurochem. 66:877-880, 1996; Poirier et al Neuroscience 55: 81-90 (1993); Cedazo-Minuez et al [Neurosci 105: 651-661, 2001; Gurwitz et al Eur. J. Pharmacol. 267, 21, 1993; Fisher et al, J. Neural Transm Suppl 62: 189, 2002; Chen et al, J Neurotrauma, 15: 231-237, 1998; Dantzer et al. Psychopharmacol. 91:363-368, 1987; Perio et al Psychopharmacol. 97: 262-268. 1989; Perio et al Psychopharmacol. 97: 262-268. 1989; Fisher et al, J. Pharmacol. Exptl. Therap., 257: 10 392, 1991; Simons et al Life Sci., 42, 375-383, 1988; Simons et al., 1988; Vincent et al Brain Res., 597, 264-268, 1992; Schwarz et al Drug Dev. Res., 40, 133-143, 1997; Voll et et al, Stroke, 20: 1700-1706, 1989; Bymaster et al. J Pharmacol Exp Ther 267: 16-24, 1993; Roldan et al Neurosci. Lett. 230: 93-96, 1997; Kimura et al Brain Res. 834: 6-12, 1999; Garrido JL et al. (FASEB J 2002; 16:1982. 15 Summary of the Invention There is provided in accordance with an embodiment of the invention a compound of the formula (I): C/A )c/a I Ib wherein: C denotes a spiro carbon atom shared by ring A and the ring containing a, b, d and e; 20 A is selected from the group consisting of: RN C RN C C C NN and wherein R is selected from H, C 1
-C
8 straight- or branched-chain alkyl, or -CH 2
-P(=O)(OH)
2 ; a is -0- or -S-; 25 b is -CRR 2 - or-C(R )=; d is selected from the group consisting of =N-, -C(=O)-, -C(=S)- and =N(R)=O; e is selected from the group consisting of -CH 2 -, -CHIR 4 -, -NH-, -NR-, -N(SO2R 6 )- and -N(C(=O)R 6)-; R, R 1 , R , R, R 4 are each independently selected from H, CI-.
6 alkyl optionally substituted by one, two 30 or three phenyls, Cl- 6 alkoxy, C 2 6 hydroxyalkyl, CI.
6 alkenyl, and C 2 .6 alkynyl;
R
s is independently selected from H, CL.6 alkyl optionally substituted by one, two or three phenyls, CL-6 alkoxy, C 2
.
6 hydroxyalkyl, C 2
.
6 alkenyl, C16 alkynyl, substituted phenyl, and heteroaryl; and WO 03/092580 PCT/IL03/00357 3
R
6 is selected from C 1
.-
6 alkyl, C 1
-
6 alkoxy, C 1 i-6 alkylthio, C2-a hydroxyalkyl, C 2
-
6 alkenyl, C 2
.
6 alkynyl, and C 3 -7 cycloalkyl, each optionally substituted by from 1-6 halogen atoms, hydroxy-CI.
6 -alkyl, aryl substituted with a halogen, nitro, amino, hydroxyl, or CF 3 group, and C 1 -6 alkyl substituted by one, two or three aryl groups, C.
6 alkyl indole, isoindolyl, 3-pyridinyl, 3-piperidinyl, benzimidazolyl, thienyl, 5 isothiazolyl, imidazolyl, pyrazinyl, benzofuranyl, quinolyl, isoquinolyl, benzothienyl, isobenzofuryl, pyrazolyl, indolyl, isoindolyl, benzimidazolyl, purinyl, carbazolyl, oxazolyl, thiazolyl, isothiazolyl, 1,2,5-thiadiazolyl, isooxazolyl, pyrrolyl, pyrazolyl, quinazolinyl, pyridazinyl, pyrazinyl, cinnolinyl, phthalazinyl, quinoxalinyl, xanthinyl, hypoxanthinyl, and pteridinyl; or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or 10 pharmaceutically acceptable salt thereof, RN C with the proviso that when A is , R is -CH 3 , a is S, b is -CH(CH 2
CH
3 )- and d is -C(=O)-, then e is not -NH- (AF267 or an enantiomer thereof), and with the RN C further proviso that when A is , R is -CH 3 , a is S, b is -C(CH 3 )= and d is =N-, then e is not -CH 2 - (AF 150(S)). 15 In an embodiment of the invention, R is heteroaryl selected from the group consisting of indole, pyrrolidinyl, piperidinyl, piperazinyl, furyl, pyridyl, pyrimidyl, thienyl, isothiazolyl, imidazolyl, pyrazinyl, benzofuranyl, quinolyl, isoquinolyl, benzothienyl, isobenzofuryl, pyrazolyl, indolyl, isoindolyl, benzimidazolyl, purinyl, carbazolyl, oxazolyl, thiazolyl, isothiazolyl, 1,2,5-thiadiazolyl, isooxazolyl, pyrrolyl, pyrazolyl, quinazolinyl, pyridazinyl, pyrazinyl, cinnolinyl, phthalazinyl, 20 quinoxalinyl, xanthinyl, hypoxanthinyl, pteridinyl, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and pyrazolopyrimidinyl. In an embodiment of the invention, the compound is a dimer of a compound of formula 1, wherein e is -NR 5 - and the two formula 1 moieties share a common group Rs which is selected from the group consisting of -(CH 2 ),- and -(CH 2 0),-, wherein n is 1 to 6, or an enantiomer, diastereomer, 25 racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof. In an embodiment of the invention, the compound is selected from the group consisting of: N-[(2,8-Dimethyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene)-amine]- N-oxide; N-[(2-Ethyl- 8-methyl 1-oxa- 8-aza-spiro[4.5]dec- 3-ylidene)- amine]-N-oxide; N-[(2-Methyl- 8-phenyl- 1-oxa- 8-aza spiro[4.5]dec- 3-ylidene)- amine]-N-oxide; Thia-4,8-diaza- spiro[4.5]decan- 3-one; 4-(2,4-Dimethoxy 30 benzyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF286); 8-Methyl- 4-pyrrolidin 1-ylmethyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF287); 2-(1-Hydroxy-ethyl)- 8-methyl- 1-thia 4,8-diaza- spiro[4.5]decan- 3-one (AF298); (S)-2-Ethyl- 8-methyl- 8-oxy- 1-thia- 4.8-diaza spiro[4.5]decan- 3-one (AF299); 4-(2,4-Dimethoxy- benzyl)- 2-ethyl- 8-methyl- 1-thia- 4,8-diaza spiro[4.5] decan- 3-one (AF288); (S)-2-Ethyl- 8-methyl- 1-oxo- 11 4 -thia- 4,8-diaza- spiro[4.5]decan 35 3-one (AF300); 2-Ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decane- 3-thione (AF510); (S)-2-Ethyl- WO 03/092580 PCT/IL03/00357 4 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF700); 2-Ethyl 4-[2-(1H-indol-3-yl)-ethyl]- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF703); 2-Ethyl 4-(3-1H-indol-3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF704); (S)-Ethyl 4-(3-1H-indol-3-yl-propionyl)-8-methyl- 1-thia- 4,8-diaza-spiro[4.5]decan-3-one (AF704B); (R)-Ethyl 5 4-(3-1H-indol-3-yl-propionyl)-8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF704A); and 2-Methyl-8-methyl-d 3 -1-thia-3,8-diaza-spiro[4.5]dec-2-ene (AF402), or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, metabolite, or a pharmaceutically acceptable salt thereof. In an embodiment of the invention, the compound is AF292 or a pharmaceutically acceptable salt 10 thereof. RN C In an embodiment of the invention, the compound is a compound wherein A is , R is H, a is -S-; b is -CH(CH 2
CH
3 )-; d is -(C=0)-; and e is -NH-, i.e. 2-Ethyl-1-thia-4,8-diaza-spiro[4.5]decan-3-one (AF504), or an enantiomer, diastereomer, geometrical isomer, racemate, tautomer, dimer, metabolite or pharmaceutically acceptable salt thereof. 15 In an embodiment of the invention, the compound is (S)-2-Ethyl- 1-thia- 4,8-diaza spiro[4.5]decan-3-one (AF292) or its HCI salt. In an embodiment of the invention, the compound is (R)-2-Ethyl- 1-thia- 4,8-diaza spiro[4.5]decan-3-one (AF291). RN C In an embodiment of the invention, the compound is a compound wherein A is , R 20 is -CH 3 , a is -0-, d is =N(R 3 )=0, or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof. In an embodiment of the invention, b is -CH(CH 3 )- and R 3 is -CH 3 , i.e. N-[(2,8-Dimethyl- 1-oxa- 8-aza- spiro[4.5] dec-3-ylidene)- methyl-amine]- N-oxide (AF600). In an embodiment of the invention, b is -CH(CH 3
)
and R is benzyl, i.e. N-[(2,8-Dimethyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene)- benzyl-amine] 25 N-oxide (AF604). In an embodiment of the invention, b is -CH(CH 3 )- and R is isopropyl, i.e. N-[(2,8-Dimethyl- 1-oxa- 8-aza- spiro[4.5] dec- 3-ylidene)- isopropyl-amine] -N-oxide (AF605). In an embodiment of the invention, b is -CH(CH 2
CH
3 )- and R is -CH 3 , i.e. N-[(2-Ethyl- 8-methyl- 1-oxa 8-aza- spiro[4.5] dec-3-ylidene)-methyl-amine]- N-oxide (AF601). In an embodiment of the invention, b is -CH(CH 3 )- and R 3 is phenyl, i.e. N-[(2-Methyl- 8-phenyl- 1-oxa- 8-aza-spiro 30 [4.5]dec-3-ylidene)-methyl-amine]-N-oxide (AF602). In an embodiment of the invention, the compound is a compound wherein A is C R is -CH 3 , a is -0-, d is =N(R)=0, or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof. In an embodiment of the WO 03/092580 PCT/IL03/00357 5 invention, b is -CH(CH 3 )- and R 3 is methyl, i.e. Dihydro- 5'-methylspiro [I-azabicyclo[2.2.2]octane 3,5'-(4'H)- 3'-ylidene- methylamnine]-N-oxide (AF603). RN C In an embodiment of the invention, the compound is a compound wherein A is , R is methyl, a is -S-, b is -CH(CH 2
CH
3 )-; d is -C(=O)-; e is -NRa- wherein R is selected from 5 -(CH 2
)
3 -indolyl and -C(=O)-(CHz) 3 -indolyl, i.e. 2-Ethyl- 4-[2-(1H-indol-3-yl)-ethyl]- 8-methyl- 1-thia 4,8-diaza- spiro[4.5]decan- 3-one (AF703) or 2-Ethyl- 4-(3-1H-indol-3-yl-propionyl)- 8-methyl- 1-thia 4,8-diaza- spiro[4.5]decan- 3-one (AF704), or an enantiomer, diastereomer, geometrical isomer, racemate, tautomer, dimer, metabolite or pharmaceutically acceptable salt thereof. In an embodiment of the invention, the is (S)-Ethyl- 4-(3-1H-indol- 3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza 10 spiro[4.5]decan-3-one (AF704B). There is also provided in accordance with an embodiment of the invention the use of a compound of formula (I), or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition. In an embodiment of the invention, the compound is AF292 or a prodrug of AF292 or a 15 pharmaceutically acceptable salt of either AF292 or a prodrug of AF292. In an embodiment of the invention, the prodrug is AF267B or a pharmaceutically acceptable salt thereof. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising at least one compound of formula (I), or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof, 20 and a pharmaceutically acceptable carrier, diluent or excipient therefor. In an embodiment of the invention, the compound is AF292 or a prodrug of AF292 or a pharmaceutically acceptable salt thereof. In an embodiment of the invention, the prodrug is AF267B or a pharmaceutically acceptable salt thereof. There is also provided in accordance with an embodiment of the invention a method of 25 stimulating the M1 muscarinic receptor, comprising administering to a patient in need thereof an efficacious amount of a compound selected from the group consisting of compounds of formula (I), AF267 and AF150(S), or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof. In an embodiment of the invention, the compound is AF292 or a prodrug of AF292 or a pharmaceutically acceptable salt thereof. In an 30 embodiment of the invention, the prodrug is AF267B or a pharmaceutically acceptable salt thereof. There is also provided in accordance with an embodiment of the invention a compound of RN C formula (I) wherein A is , R is -CH 3 , a is -0-, d is =N(R 3 )=0, or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof in the preparation of a pharmaceutical composition for both stimulating the M1 35 muscarinic receptor and retarding oxidation in the vicinity of said M1 muscarinic receptor.
WO 03/092580 PCT/IL03/00357 6 There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising an Ml muscarinic receptor agonistic efficacious amount of a compound of RN C formula (I) wherein A is , R is -CH 3 , a is -0-, d is =N(R)=O, or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically 5 acceptable salt thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. There is also provided in accordance with an embodiment of the invention a method of stimulating the M1 muscarinic receptor, comprising administering to a patient in need thereof an RN C efficacious amount of a compound of formula (I) wherein A is , R is -CH 3 , a is -0-, d is 10 =N(R 3 )=0, or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof. There is also provided in an embodiment of the invention the compound (S)-2-Ethyl 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF700). There is also provided in accordance with an embodiment of the invention the use of the 15 compound AF700, or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition for both stimulating the M1 muscarinic receptor and activating cc secretase. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising an M1 muscarinic receptor stimulating and a- secretase activating efficacious 20 amount the compound AF700, or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. There is also provided in accordance with an embodiment of the invention a method of stimulating the M1 muscarinic receptor and activating ca secretase, comprising administering to a 25 patient in need thereof an efficacious amount of the compound AF700, or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof. There is also provided in accordance with an embodiment of the invention the use of the compound AF700, or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition for both stimulating the M1 30 muscarinic receptor and antagonizing P3 secretase. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising an Ml muscarinic receptor agonistic and P-secretase antagonistic efficacious amount of the compound AF700, or an enantiomer, diastereomer, geometrical isomer, racemate, tautomer, dimer, metabolite or pharmaceutically acceptable salt thereof, and at least one 35 pharmaceutically acceptable carrier, diluent or excipient therefor.
WO 03/092580 PCT/IL03/00357 7 There is also provided in accordance with an embodiment of the invention a method of stimulating the M1 muscarinic receptor and antagonizing 3-secretase, comprising administering to a patient in need thereof an efficacious amount of the compound AF700, or an enantiomer, diastereomer, geometrical isomer, racemate, tautomer, dimer, metabolite or pharmaceutically acceptable salt thereof. 5 There is also provided in accordance with an embodiment of the invention the use of the compound AF700, or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition for both stimulating the M1 muscarinic receptor and antagonizing y-secretase. There is also provided in accordance with an embodiment of the invention a pharmaceutical 10 composition comprising an M1 muscarinic receptor agonistic and y-secretase antagonistic efficacious amount of the compound AF700, or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. There is also provided in accordance with an embodiment of the invention a method of 15 stimulating the M1 muscarinic receptor and antagonizing y-secretase, comprising administering to a patient in need thereof an efficacious amount of the compound AF700, or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof. There is also provided in accordance with an embodiment of the invention the use of the compound (S)-2-Ethyl-l1-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292), or a metabolite or 20 pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition for both stimulating the M1 muscarinic receptor and antagonizing the M3 muscarinic receptor. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising an M1 muscarinic receptor agonistic and M3 muscarinic receptor antagonistic efficacious amount of the compound (S)-2-Ethyl-l-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292), or a 25 metabolite or pharmaceutically acceptable salt thereof, and at least one pharmaecutically acceptable carrier, diluent or excipient therefor. There is also provided in accordance with an embodiment of the invention human or animal blood containing the compound (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF292), or a metabolite or pharmaceutically acceptable salt thereof. In an embodiment of the invention, the blood is 30 located in a human or animal body. In an embodiment of the invention, the blood is not located in a human or animal body. There is also provided in accordance with an embodiment of the invention human or animal blood plasma containing the compound (S)-2-Ethyl-l-thia- 4,8-diaza-spiro[4.5] decan-3-one (AF292), or a metabolite or pharmaceutically acceptable salt thereof. In an embodiment of the invention, the 35 blood is located in a human or animal body. In an embodiment of the invention, the blood is not located in a human or animal body. There is also provided in accordance with an embodiment of the invention the compound (S)-2-Ethyl-l-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292) according to claim 1, or a metabolite or pharmaceutically acceptable salt thereof, whenever located in a human or animal body.
WO 03/092580 PCT/IL03/00357 8 There is also provided in accordance with an embodiment of the invention the compound (S) 2-ethyl-8-methyl-1-thia-4,8-diaza-spiro[4.5]decan-3-one (AF267B), for use as a prodrug of the compound (S)-2-Ethyl-l1-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292). There is also provided in accordance with an embodiment of the invention the use of the 5 compound (S)-2-ethyl-8-methyl-l-thia- 4,8-diaza- spiro[4.5] decan- 3-one (AF267B) as a prodrug of the compound (S)-2-Ethyl-l1-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292). There is also provided in accordance with an embodiment of the invention the use of a combination of the compounds (S)-2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF267B) and (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5J decan-3-one (AF292) in the preparation of a 10 pharmaceutical composition for stimulating the Ml muscarinic receptor and antagonizing the M3 muscarinic receptor. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising an M1 muscarinic receptor agonistic and M3 muscarinic receptor antagonistic amount of a combination of the compounds AF267B and AF292 and at least one pharmaceutically 15 acceptable carrier, diluent or excipient therefor. There is also provided in accordance with an embodiment of the invention a method of stimulating the M1 muscarinic receptor and antagonizing the M3 muscarinic receptor in a patient, comprising administering to a patient an efficacious amount of a combination of the compounds AF267B and AF292. 20 In an embodiment of the invention, AF267B and AF292 are administered together. In an embodiment of the invention, AF267B and AF292 are administered separately. In an embodiment of the invention, AF267B and AF292 are administered at different times. In an embodiment of the invention, AF267B and AF292 are administered at the same times. There is also provided in accordance with an embodiment of the invention the use of a 25 combination of a first compound which is (S)-2-Ethyl- 1-thia- 4,8-diaza-spiro[4.5] decan-3-one (AF292) and a second compound selected from the group consisting of compound of formula (I), AF267B and AF 150(S), including racemates, enantiomers, diastereomers, tautomers, geometric isomers and pharmaceutically acceptable salts thereof, in the preparation of a pharmaceutical composition for stimulating the M1 muscarinic receptor while minimizing adverse side-effects due to stimulation of 30 other mAChIR subtypes. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising an M1 muscarinic receptor agonistic amount of a combination of a first compound which is AF292 and a second compound selected from the group consisting of compounds of formula (I), AF267B and AF150(S), including racemates, enantiomers, diastereomers, geometric 35 isomers, tautomers and pharmaceutically acceptable salts thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. There is also provided in accordance with an embodiment of the invention a method of stimulating the M1 muscarinic receptor while minimizing adverse side-effects due to stimulation of other mAChR subtypes in a patient, comprising administering to a patient an efficacious amount of a WO 03/092580 PCT/IL03/00357 9 combination of a first compound which is AF292 and a second compound selected from the group consisting of compounds of formula (I), AF267B and AF150(S), including racemates, enantiomers, diastereomers, tautomers, geometric isomers and pharmaceutically acceptable salts thereof. In an embodiment of the invention, the first compound and the second compound are administered together. 5 In an embodiment of the invention, the first compound and the second compound are administered separately. In an embodiment of the invention, the first compound and the second compound are administered at different times. In an embodiment of the invention, the first compound and the second compound are administered at the same time. There is also provided in accordance with an embodiment of the invention a method for 10 stimulating the M1 muscarinic receptor in a patient simultaneously with AF267B and AF292, comprising administering to a patient an amount of AF267B efficacious to form in vivo an amount of a mixture of AF267B and AF292 efficacious to stimluate the Ml muscarinic receptor. There is also provided in accordance with an embodiment of the invention 2-Ethyl 4-(3-1H-indol- 3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan- 3-one in racemic form 15 (AF704) or as the S-enantiomer thereof (AF704B), for use as a prodrug of at least one of the group of AF267B, AF292 and indole-3-propionic acid. There is also provided in accordance with an embodiment of the invention the use of 2-Ethyl-4-(3-1H-indol-3-yl-propionyl)-8-methyl- 1-thia-4,8-diaza- spiro[4.5] decan-3-one in racemic form (AF704) or as the S-enantiomer thereof (AF704B), in the preparation of a pharmaceutical 20 composition for stimulating the Ml muscarinic receptor, retarding oxidation in the vicinity of the M1 muscarinic receptor, and providing neuroprotectant activity. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising an M1 muscarinic receptor stimulating, oxidation-retarding and neuroprotectant activity efficacious amount of the compound 2-Ethyl- 4-(3-1H-indol- 3-yl-propionyl) 25 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one in racemic form (AF704) or as the S-enantiomer thereof (AF704B), and a pharmaceutically acceptable carrier, diluent, or excipient therefor. There is also provided in accordance with an embodiment of the invention a process for the preparation of 2-ethyl-8-methyl-l1-thia-4,8-diaza- spiro[4.5]decan-3-one (AF267), comprising reacting 4-ethyl piperidone with 2-mercaptobutyric acid and ammonia. In an embodiment of the invention, the 30 process further comprising obtaining the enantiomers AF267A (R-enantiomer) and AF267B (S-enantiomer) by chiral separation. There is also provided in accordance with an embodiment of the invention a process for the preparation of AF267B, comprising racemizing AF267A and isolating AF267B from the racemic mixture. In an embodiment of the invention, the isolating comprising separating the AF267B from the 35 racemic mixture by chiral separation. There is also provided in accordance with an embodiment of the invention a process for the synthesis of AF267B comprising contacting (R)-2-mercaptobutyric acid with ammonium acetate and 1-methyl-4-piperidone. In an embodiment of the invention the (R)-2-mercaptobutyric is obtained by contacting (R)-2-benzoylthiobutyric acid with ammonium hydroxide. In an embodiment of the WO 03/092580 PCT/IL03/00357 10 invention the (R)-2-benzoylthiobutyric acid is obtained by contacting (R)-2-bromobutyric acid with cesium thiobenzoate. In an embodiment of the invention the (R)-2-bromrnobutyric acid is obtained by contacting 2-aminobutyric acid having the R configuration with sodium nitrite, potassium bromide and hydrobromic acid. 5 There is also provided in accordance with an embodiment of the invention a process for the preparation of 1 4 C-labelled AF267B, comprising reacting AF287 with 14 C-labelled ethyl bromide, deprotecting the nitrogen atom at the 4-position of the AF287, and isolating 1 4 C-labelled AF267B by chiral chromatography. There is also provided in accordance with an embodiment of the invention a process for the 10 preparation of a mixture of (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF292) and (R)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF291) comprising reacting AF267 with a demethylating agent. There is also provided in accordance with an embodiment of the invention a crystalline form of (S)-2-ethyl-8-methyl-l1-thia-4,8-diaza- spiro[4.5]decane -3-one (AF267B) characterized by the 15 following data: P212121 (No 16) a=10.394 ((10) (ct=90o), b=20.133 (2) (3=90o), c=5.856 (4) (y=-90'), A, T=110K. In an embodiment of the invention, the crystalline form is further characterized by the following data: Volume=1224.2 (9) A 3 , Z=4, Fw=202.32, Calculated density, Dc=1.092Mg/mn 3 , Absorption coefficient, .t =0.232mm " 1 . There is also provided in accordance with an embodiment of the invention a crystalline form of 20 (AF267B) having the configuration shown in Structure 1 in Example 2. There is also provided in accordance with an embodiment of the invention a process for the preparation of 2,8-Dimethyl- 1-thia- 3,8-diaza- spiro[4.5] dec- 2-ene [A]150(S)] comprising cyclizing 1-methyl- 4-N-thioacetylamino- 1,2,3,6-tetra- hydropyridine. In an embodiment of the invention the cyclizing is conducted in the presence of phosphoric acid. In an embodiment of the invention the 1 25 methyl- 4-N-thioacetylamino- 1,2,3,6-tetrahydropyridine is obtained by reduction of reduction of 1-methyl- 4-N-thioacetylaminomethyl pyridinium with sodium borohydride. In an embodiment of the invention the 1-methyl-4-N-thioacetylaminomethyl pyridinium is obtained by reacting 4-(acetaminomethyl)-l-methlyl-pyridinium iodide with Lawesson's reagent. There is also provided in accordance with an embodiment of the invention apharmaceutical 30 formulation comprising AF150(S) in paraffin oil. There is also provided in accordance with an embodiment of the invention a method for inhibiting the release or synthesis of beta-amyloid peptide (AP3) in a mammalian cell, tissue or organism comprising administering to a mammalian cell, tissue or organism an amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B 35 and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to inhibit the cellular release or synthesis of AP3. There is also provided in accordance with an embodiment of the invention the use of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AFl50(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and WO 03/092580 PCT/IL03/00357 11 pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for inhibiting the release or synthesis of beta-amyloid peptide (AP3) in a mammalian cell, tissue or organism. There is also provided in accordance with an embodiment of the invention a method for elevating 5 the level of secreted form of the non-amyloidogenic amyloid precursor protein (cx-APPs) in a mammalian cell, tissue or organism comprising administering to a mammalian cell, tissue or organism an amount of a compound or a mixture of compounds selected from the group consisting of compound of formula (I), AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to elevate the level of the secreted 10 form of the non-amyloidogenic amyloid precursor protein (a-APPs). There is also provided in accordance with an embodiment of the invention the use of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts, in the preparation of a pharmaceutical composition for elevating the 15 level of secreted form of the non-amyloidogenic amyloid precursor protein (c.-APPs) in a mammalian cell, tissue or organism. There is also provided in accordance with an embodiment of the invention a method for decreasing the level of AP3 peptide in the brain of a mammal having an elevated level of A3 in the brain, comprising administering to a mammal having an elevated level of AP in the brain an amount of a 20 compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to decrease the level of AP3 in the brain of said mammal. In an embodiment of the invention the elevated level of AP3 in the brain is a result of hypercholesterolemia. In an embodiment of the invention the elevated level of A3 in the brain is a 25 result of cholinergic hypofunction. There is also provided in accordance with an embodiment of the invention the use of a compound or a mixture of compounds selected from the group consisting of compound of formula (I), AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for 30 decreasing the level of AP peptide in the brain of a mammal having an elevated level of A3 in the brain. There is also provided in accordance with an embodiment of the invention a method for inhibiting the synthesis or release of apolipoprotein (ApoE) in a mammalin cell, tissue or organism comprising administering to a mammalian cell, tissue or organism an amount of a compound or a mixture of compounds selected from the group consisting of compound of formula (I), AF267B and 35 AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to inhibit the release or synthesis of ApoE in said mammalian cell, tissue or organism. There is also provided in accordance with an embodiment of the invention the use of a compound or a mixture of compounds selected from the group consisting of compound of formula (I), AF267 and WO 03/092580 PCT/IL03/00357 12 AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for inhibiting the release or synthesis of ApoE in a mammalian cell, tissue or organism. In an embodiment of the invention the ApoE is ApoE4. 5 There is also provided in accordance with an embodiment of the invention a method for decreasing levels of apolipoprotein (ApoE) in a mammalian cell, tissue or organism comprising administering to a mammalian cell, tissue or organism an amount of a compound or a mixture of compounds selected from the group consisting of compound of formula (I), AF267B and AF150(S), or racemates, geometrical isomers, enantiomers, diasteromers, tautomers and pharmaceutically acceptable 10 salts thereof effective to decrease the levels of ApoE in said mammalian cell, tissue or organism. There is also provided in accordance with an embodiment of the invention the use of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for 15 decreasing levels of ApoE in a mammalian cell, tissue or organism. In an embodiment of the invention the ApoE is ApoE4. There is also provided in accordance with an embodiment of the invention a method for decreasing tau hyperphosphorylation in a mammalian cell, tissue or organism comprising administering to a mammalian cell, tissue or organism a compound or a mixture of compounds selected from the 20 group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to inhibit tau hyperphosphorylation. In an embodiment of the inventionthe tau hyperphosphorylation is A3-induced tau hyperphosphorylation. There is also provided in accordance with an embodiment of the invention the use of a compound 25 or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for decreasing tau hyperphosphorylation in a mammalian cell, tissue or organism. There is also provided in accordance with an embodiment of the invention a method for 30 decreasing paired helical formation in a mammalian cell, tissue or organism comprising administering to a mammalian cell, tissue or organism a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to inhibit tau hyperphosphorylation. 35 There is also provided in accordance with an embodiment of the invention the use of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267 and AF150(S), or racemates, enantiomers, diasteromers, tautomers, geometrical isomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for decreasing paired helical formation in a mammalian cell.
WO 03/092580 PCT/IL03/00357 13 There is also provided in accordance with an embodiment of the invention a method for activating the Wnt signaling pathway in a mammalian cell, tissue or organism comprising administering to a mammalian cell, tissue or organism a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, 5 diasteromers, tautomers, geometrical isomers and pharmaceutically acceptable salts thereof effective to inhibit Wnt abnormalities. There is also provided in accordance with an embodiment of the invention the use of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267 and AF150(S), or racemates, enantiomers, diasteromers, tautomers, geometrical isomers and 10 pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for activating the Wnt signaling pathway in a mammalian cell, tissue or organism. There is also provided in accordance with an embodiment of the invention a method for inhibiting GSK3P3-mediated effects in a mammalian cell, tissue or organism comprising administering to a mammalian cell, tissue or organism a compound or a mixture of compounds selected from the 15 group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers, geometrical isomers and pharmaceutically acceptable salts thereof effective to inhibit GSK3 3-mediated effects. In an embodiment of the invention the GSK3 P3-mediated effects are selected from the group consisting of tau hyperphosphorylation, apoptosis, P-catenin degradation, and decrease in Wnt target genes. 20 There is also provided in accordance with an embodiment of the invention the use of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267 and AF150(S), or racemates, enantiomers, diasteromers, tautomers, geometrical isomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for inhibiting GSK3P3-mediated effects in a mammal. In an embodiment of the invention the method is 25 used in response to insults induced by AP3 peptides or oxidative stress starvation to Wnt signaling, apoptosis, or cell viability. There is also provided in accordance with an embodiment of the invention a method for enhancing the activity of endogenous growth factors, i.e. neutrophins, in a cell, comprising administering to a mammalian cell, tissue or organism an amount of a compound or a mixture of 30 compounds selected from the group consisting of compounds of formula (I), AF267B and AF 150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof which alone is effective as a neurotrophic agent and which acts synergistically with said neurotrophins. There is also provided in accordance with an embodiment of the invention the use of a compound 35 or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers, geometrical isomers and pharmaceutically acceptable salts thereof which alone is effective as a neurotrophic agent and which acts synergistically with endogenous growth factors, i.e. neurotrophins, in the preparation of a pharmaceutical composition for enhancing the activity of neurotrophins.
WO 03/092580 PCT/IL03/00357 14 There is also provided in accordance with an embodiment of the invention a pharmaceutical composition for inhibiting the release or synthesis of beta-amyloid peptide (AP3), for elevating the level of secreted form of the non-amyloidogenic amyloid precursor protein (c-APPs), for decreasing the level of AP peptide in the brain of a mammal having an elevated level of AP in the brain, for inhibiting 5 the release or synthesis of ApoE, for decreasing levels of ApoE, for decreasing tau hyperphosphorylation, for decreasing paired helical formation, for activating the Wnt signaling pathway, for increasing beta-catenin, for inhibiting GSK3P-mediated effects or for for enhancing the activity of neurotrophins, comprising a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, 10 geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising at least one compound selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, 15 tautomers and pharmaceutically acceptable salts thereof and at least one additional pharmacologically active compound selected from the group constisting of: cholinesterase inhibitors, nicotinic agonists, cholinergic precursors and cholinergic enhancers, nootropics, peripheral antimuscarinc drugs, M2 muscarinic antagonists, M4 antagonists, benzodiapine inverse agonists, antidepressants, tricyclic antidepressents or antimuscarinic drugs used in treatment of Parkinson's disease (PD) or depression, 20 antipsychotic and antischizophrenic agents, glutamate antagonists and modulators, NMDA antagonists, AMPA agonists, acetyl-L-camitine, MAO-B inhibitors, peptides and growth factors, cholesterol-lowering agents, antioxidants, GSK-313 inhibitors, Wnt-ligands, 3- or y-secretase inhibitors, beta-amyloid degrading agents, beta-amyloid anti-aggregation agents, chelating agents, immunotherapeutic compounds against beta-amyloids, compounds that bind to amyloids, 25 cyclooxygenase (COX)-2 inhibitors, non-steroidal antiinflammnatory drugs, estrogenic agents, estrogenic receptor modulators, steroidal neuroprotectants, and spin trapping pharmaceuticals. In an embodiment of the invention the compound is AF292 or a prodrug of AF292 or a pharmaceutically acceptable salt thereof. There is also provided in accordance with an embodiment of the invention a method for treating 30 or reducing cerebral amyloid angiopathy comprising administering to a patient in need thereof (a) an efficacious amount of a compound selected from the group consisting of AF267B, AF292, and AF704B pharmaceutically acceptable salts thereof or mixtures of such compounds or salts, and (b) an efficacious amount of a compound selected from an immunotherapeutic compound against beta-amyloids and compounds that bind to amyloids. 35 There is also provided in accordance with an embodiment of the invention a pharmaceutical composition comprising (a) an efficacious amount of a compound selected from the group consisting of AF267B, AF292, and AF704B pharmaceutically acceptable salts thereof or mixtures of such compounds or salts, and (b) an efficacious amount of a compound selected from an immunotherapeutic WO 03/092580 PCT/IL03/00357 15 compound against beta-amyloids and compounds that bind to amyloids, and a pharmaceutically acceptable carrier, diluent or exicipient therefor. There is also provided in accordance with an embodiment of the invention the use of a combination of (a) a compound selected from the group consisting of AF267B, AF292, and AF704B 5 pharmaceutically acceptable salts thereof or mixtures of such compounds or salts, and (b) a compound selected from an immunotherapeutic compound against beta-amyloids and compounds that bind to amyloids, in the preparation of a pharmaceutical composition for treating or reducing cerebral amyloid angiopathy. There is also provided in accordance with an embodiment of the invention a method for treating 10 in a mammal diseases associated with impaired cholinergic function or diseases where there is an imbalance in cholinergic function, or diseases with impared activity of acetylcholine receptors from the group consisting of: senile dementia of Alzheimer's type; Alzheimer's disease (AD); Lewy body dementia; mixed Alzheimer's and Parkinson's disease; multiifract dementia (MID); fronto-temporal dementia; vascular dementia; stroke/ischemia, MID combined with stroke/ischemia/head injury; 15 combined MID and AD; human head injury; age-associated memory impairments; mild cognitive impairment (MCI); MCI conducive to AD; cognitive dysfunction (including forgetfulness, acute confusion disorders, attention-deficit disorders, focus and concentration disorders); hallucinatory-paranoid states; emotional and attention disorders; sleep disorders; post-operative delirium; adverse effects of tricyclic antidepressants; adverse effects of certain drugs used in the 20 treatment of schizophrenia and Parkinson's disease; xerostomia, anomia, memory loss and/or confusion; psychosis; schizophrenia, schizophrenia comorbit with AD, late onset schizophrenia, paraphrenia, schizophreniform disorders; anxiety; bipolar disorders; mania; mood stabilization; cognitive impairments after removal of certain gliomas; tardive dyskinesia; oxidative stress during oxygen therapy; aphasia; postencephalitic amnesic syndrome; AIDS dementia; memory impairments in 25 autoimmune diseases including lupus, multiple sclerosis, Sjogren's syndrome, chronic fatigue syndrome, and fibromyalgia; memory impairments in atypical depression or schizophrenia; pain, rheumatism, arthritis and terminal illness;, xerophtalmia, vaginal dryness, skin dryness; immune dysfunctions; neurocrine disorders and dysregulation of food intake, including bulimia and anorexia; obesity; congenital ornithine transcarbamylase deficiency; ollivopontocerebral atrophy; alcohol 30 withdrawal symptoms; substance abuse including withdrawal symptoms and substitution therapy; Huntington's chorea; progressive supranuclear palsy; Pick's disease; Friedrick's ataxia; Gilles de la Tourette disease; Down's syndrome; glaucoma; presbyopia; autonomic disorders including dysfunction of gastrointestinal motility and function such as inflammatory bowel disease, irritable bowel syndrome, diarrhea, constipation, gastric acid secretion and ulcers; urinary urge incontinence, asthma, COPD; 35 comprising administering to a mammal in need of such treatment a compound a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in an amount effective to treat one of said diseases. In an embodiment of the invention the compound is AF292 or a prodrug of AF292 or a pharmaceutically acceptable salt thereof.
WO 03/092580 PCT/IL03/00357 16 There is also provided in accordance with an embodiment of the invention a method for preventing or treating central or peripheral nervous system disease states due to dysfunction in one or more of the following: brain, nervous system, cardiovascular system, immune system, neurocrine system, gastrointestinal system, or endocrine and exocrine glands, eye, cornea, lungs, prostate, or other 5 organs where the cholinergic function is mediated by muscarinic receptor subtypes, wherein said dysfunction involves: brain amyloid-mediated disorders; glycogen synthase kinase (GSK3 3)-mediated disorders; tau protein hyperphosphorylation-mediated damages, dysfunctions or diseases; CNS and PNS hypercholesterolemia- and/or hyperlipidemnia-mediated damages, dysfunctions or diseases; Wnt-mediated signaling abnormalities; impairment of neuroplasticity; hyperglycemia; diabetes; 10 endogenous growth factors-mediated diseases, or combination of additional risk factors; or disease states that involve apolipoprotein E; or disturbances in which a cholinergic dysfunction has been implicated, including: senile dementia of Alzheimer's type, Alzheimer's disease (AD), delay of onset of AD symptoms in a patient at risk for developing AD, Lewy body dementia, cerebral amyloid angiopathy (CAA), cerebral amyloidosis, fronto-temporal dementia, vascular dementia, hyperlipidemia, 15 hypercholesterolemia, multiifract dementia (MID), stroke ischemia, MID combined with stroke/ischemia/head injury, combined MID and Alzheimer's disease, human head injury, age-associated memory impairments, mild cognitive impairment (MCI), MCI conducive to AD, bipolar disorder, mania, schizophrenia, nonaffective sychozophrenia, paraphrenia, immune dysfunctions, neurocrine disorders and dysregulation of food intake, including bulimia and anorexia, weight control, 20 obesity, inflammation; comprising administering to a mammal in need of such treatment a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in an amount effective to treat at least one of said diseases. In an embodiment of the invention the compound is AF292 or a prodrug of AF292 or a pharmaceutically acceptable salt thereof. 25 There is also provided in accordance with an embodiment of the invention a method for treating a patient with AD, MCI, Lewi Body Dementia, fronto-temporal dementia, vascular dementia, memory impairment in head injury, AIDS dementia in order to inhibit further deterioration in the condition of said patient comprising administering to said patient an efficacious amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I), AF267B and AF150(S), 30 or racemates, enantiomers, diasteromers, tautomers, geometric isomers and pharmaceutically acceptable salts thereof. In an embodiment of the invention the compound is AF292 or a prodrug of AF292 or a pharmaceutically acceptable salt thereof. There is also provided in accordance with an embodiment of the invention a method of treating 35 schizophrenia, comprising administering to a patient in need thereof an efficacious amount of a compound selected from the group consisting of AF267B, AF292, pharmaceutically acceptable salts thereof and mixtures of AF267B, AF292 or salts thereof. There is also provided in accordance with an embodiment of the invention the use of AF267B, AF292, pharmaceutically acceptable salts thereof or a mixture of AF267B, AF292 or pharmaceutically WO 03/092580 PCT/IL03/00357 17 acceptable salts thereof in the preparation of a pharmaceutical composition for the treatment of schizophrenia. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition for the treatment of schizophrenia, comprising AF267B, AF292, pharmaceutically 5 acceptable salts thereof or a mixture of AF267B, AF292 or pharmaceutically salts thereof and at least one pharmaceutically acceptable carrier, diluent or excipient. There is also provided in accordance with an embodiment of the invention a method of ameliorating symptoms of schizophrenia, comprising administering to a patient in need thereof an efficacious amount of a compound selected from the group consisting of AF267B, AF292, 10 pharmaceutically acceptable salts thereof and mixtures of of AF267B, AF292 or pharmaceutically acceptable salts thereof. There is also provided in accordance with an embodiment of the invention the use of AF267B, AF292, pharmaceutically acceptable salts or mixtures of AF267B, AF292 or pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for amelioration of 15 symptoms of schizophrenia. There is also provided in accordance with an embodiment of the invention a pharmaceutical composition for amelioration of symptoms of schizophrenia, comprising a compound selected from AF267B, AF292, pharmaceutically acceptable salts thereof and mixtures of AF267B, AF292 or pharmaceutically acceptable salts thereof and at least one pharmaceutically acceptable carrier, diluent 20 or excipient. In this patent application, "alkyl" means a linear or branched chain of 1-8 carbon atoms, e.g. methyl, ethyl, propyl, isopropyl etc. "alkoxy" means -O-alkyl, e.g. to methoxy, ethoxy, propxy, isopropoxy, etc. 25 "alkenyl" means a linear or branched chain of 2-8 carbon atoms having at least one C-C double bond in the chain. "alkynyl" means a linear or branched chain of 2-8 carbon atoms having at least one C-C triple bond in the chain. "alkylthio" means -S-alkyl, e.g. methylthio, ethylthio, propylthio, isopropylthio etc. 30 "cycloalkyl" refers to mono- and bicyclic ring structures containing 3-12 carbon atoms. Examples of cycloalkyl are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, decalinyl, and norbornyl. "aryl" refers to a mono- or bicyclic aromatic ring structure containing 5-12 carbon atoms. Examples of aryl are phenyl, naphthyl and benzyl. "heterocyclic" refers to mono- and bicyclic ring structures containing 4-12 carbon atoms and at 35 least one nitrogen, oxygen or sulfur atom. "heteroaryl" refers to a mono- or bicyclic aromatic ring structure containing 4-12 carbon atoms and at least one nitrogen, oxygen or sulfur atom. Examples of heterocyclic and heteraryl are indolyl, isoindolyl, 3-pyridinyl, 3-piperidinyl, benzimidazolyl, thienyl, isothiazolyl, imidazolyl, pyrazinyl, benzofuranyl, quinolyl, isoquinolyl, benzothienyl, isobenzofuryl, pyrazolyl, indolyl, isoindolyl, WO 03/092580 PCT/IL03/00357 18 benzimidazolyl, purinyl, carbazolyl, oxazolyl, thiazolyl, isothiazolyl, 1,2,5-thiadiazolyl, isooxazolyl, pyrrolyl, pyrazolyl, quinazolinyl, pyridazinyl, pyrazinyl, cinnolinyl, phthalazinyl, quinoxalinyl, xanthinyl, hypoxanthinyl, and pteridinyl, pyrrolidinyl, piperidinyl, piperazinyl, furyl, pyridyl, pyrimidyl, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and 5 pyrazolo- pyrimidinyl. "halogen atom" may be one of fluorine, chloride, bromine and iodine. Unless noted otherwise, the following abbreviations are used: AP3 3-amyloid AA arachidonic acid AAMI age associated memory impairment ACh acetylcholine AchE-Is acetyleholinesterase inhibitors AD Alzheimer's disease AGP Human c-glycoprotein AGP c-glycoprotein ApoE apolipoprotein APP amyloid precursor protein AUC area under the curve BDNF brain derived growth factor bFGF basic fibroblast growth factor CAA cerebral amyloid angiopathy CCh carbachol CDX methyl-B-cyclodextrin CE collision energy CHII closed head injury CNS central nervous system CSF cerebrospinal fluid DAPI 4,6-diamidino-2-phenylindole DCC dicyclohexylcarbodiimide DDW double distilled water DMF N,N-dimethyl formamide DMAP 4-dimethylaminopyridine DMVIPU N,N'-dimethyl-N,N'-propylene urea ECG electrocardiogram EGF epidermal growth factor FACS Fluorescence activated cell sorter FCS fetal calf serum GC gas chromatography GSK3P3 glycogen synthase kinase WO 03/092580 PCT/11L03/00357 19 HERG human ether-a-go-go related gene HPLC high performance liquid chromatograph HS horse serum HSA human serum albumin i.e.v., icy intracerebroventricular i.p., ip intraperitoneally i.v., iv intravenous LBD Lewy Body disease LC liquid chromatogrph LDA di-isopropylamine Li lithium Ml mAChR 1 muscarinic receptor mAChR muscarinic receptor mCPBA m-chloroperbenzoic acid MCI minimal cognitive impairment MID multiifract dementia MVIRSA muscarinic receptor selective agonists and potent antioxidants MS mass spectrometry MTBE Methyl-t-butyl ether MTT 3-(4,5-dimethylthiazol-2-yl) 2,5-diphenyl-tetrazolium bromide MWM Morris water maze nbm nucleus basalis magnocellularis NFT neurofibrillary tangles NGF Nerve Growth Factor NMDA N-methyl-D-Aspartate NMR nuclear magnetic resonance NOAEL no-adverse-effect-level NSS neurological severity scores OXO-M oxotremorine-M PA passive avoidance PBS phosphate-buffered saline PD Parkinson's disease PHF paired helical filaments PI phosphoinositide PKC protein kinase C PMSF Phenylmethylsulfonylfluoride PNS peripheral nervous system WO 03/092580 PCT/IL03/00357 20 po per os, oral PS-1 presenilin- 1 PZ pirenzepine QNB quinuclidinyl benzilate REL ratio of escape latency RID Ratio of Investigation Duration ROS reactive oxygen species RPL ratio of path length RSA Receptor Selective Antioxidants s.c., so subcutaneously SDAT senile dementia of Alzheimer's type SDS-PAGE sodium dodecyl sulfate-polyacrylamide gel electrophoresis SMB Simulated Moving Bed TBI traumatic brain injury Tg transgenic THF tetrahydrofuran TUNEL Terminal deoxynucleotidyl transferase (TdT) mediated dUTP nick end labeling Al (human); A2A (human); A3 (human) adenosine receptor subtypes AT1 (Human Recombinant) angiotensin BZD (central) benzodiazepine B2 (Human Recombinant) bradykinin CCKA (Human Recombinant) (CCK1) cholecystokinin D1 (Human Recombinant); D2S (Human dopamine receptor subtypes Recombinant) ETA (Human Recombinant) endothelin GABA (non-selective) gamma-aminobutyric acid GAL2 (h) galanin IL-8B (Human Recombinant) (CXCR2) chemokine receptor subtype CCR1 (Human Recombinant) chemokine receptor subtype H1 (central); H2 histamine receptor subtypes MC4 (Human Recombinant) melanocortine ML1 melatonin NK2 (Human Recombinant); NK3 (Human tachykinin Recombinant) Y1 (human); neuropeptide, Y2 (human) neuropeptide NTI (Human Recombinant) (NTS 1) neurotensin delta 2 (Human Recombinant); (DOP); opiate receptor subtypes WO 03/092580 PCT/ILO3/00357 21 kapp (KOP); opiate, mu (Human Recombinant) (MOP) ORL1 (Human Recombinant) (NOP) orphanin 5-HTIA (Human Recombinant); 5-HTIB; serotonin sutypes 5-HT 2 A (Human Recombinant); 5-HT 3 Human Recombinant); 5-HTA (Human Recombinant) (5-ht5A); 5-HT 6 (Human Recombinant); 5-HT 7 (human) sst (non-selective) somatostatin VIP1 (human) (VPAC1) vasoactive intestinal peptide V 1 a (Human Recombinant) vasopressin NE transporter (human) norepinephrine The term "geometrical isomers" refers to isomerism across a double-bond, e.g. cis/trans isomerism and E/Z isomerism, as well as conformational isomers, e.g. and syn/anti isomerism. The term "pharmaceutically acceptable addition salts" refers to salts known in the art to be acceptable in pharmaceutical practice, for example acid addition salts such as hydrochloric acid salts, 5 maleic acid salts, and citric acid salts. Pharmaceutically acceptable acid addition salts include salts derived form inorganic acids such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorus, and the like, as well as the salts derived from organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. Such salts thus include sulfate, 10 pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, phthalate, benzenesulfonate, toluenesulfonate, phenylacetate, citrate, lactate, maleate, tartrate, methanesulfonate, and the like. Also 15 contemplated are the salts of amino acids such as arginate, gluconate, galacturonate, and the like; see, for example, Berge et al., "Pharmaceutical Salts," J. of Pharmaceutical Science, 1977;66:1-19. The acid addition salts of the basic compounds are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt in the conventional manner. The free base form may be regenerated by contacting the salt form with a base and isolating the free base in the 20 conventional manner. The free base forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free base for purposes of the present invention. Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metal hydroxides, or of organic amines. Examples of metals used as cations are 25 sodium, potassium, magnesium, calcium, and the like. Examples of suitable amines are N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine, and procaine; see, for example, Berge et al., supra., 1977.
WO 03/092580 PCT/IL03/00357 22 The base addition salts of acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. The free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in a conventional manner. The free acid forms differ from their respective salt forms somewhat in certain 5 physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acid for purposes of the present invention. The term "metabolite" refers to a form of a compound obtained in a human or animal body by action of the body on the administered form of the compound, for example a de-methylated analogue of a compound bearing a methyl group which is obtained in the body after administration of the 10 methylated compound as a result of action by the body on the methylated compound. Metabolites may themselves have biological activity. The term "prodrug" refers to a form a compound which after administration to a human or animal body is converted chemically or biochemically to a different compound in said body having biological activity. A prodrug form of a compound may itself have biological activity. 15 The novel compounds of embodiments of the present invention, and compounds which may be used in accordance with embodiments of the present invention may have at least one chiral center, and may accordingly exist as enantiomers or as mixtures of enantiomers (e.g., racemic mixtures). Where the compounds possess two or more chiral centers, they may additionally exist as diastereoisomers. In some embodiments of the present invention, there are provided pharmaceutical compositions 20 and the use of certain compounds in the manufacture of pharmaceutical compositions. Such compositions may be in a form suitable for oral (e.g. in the form of capsules, tablets, granules, powders or beads), rectal, parenteral, intravenous, intradermal, subcutaneous, transdermal or topical administration, or for administration by insufflation or nasal spray, iontophoretic, bucal, or sublingual lingual administration. Such compositions may be in unit dosage form. The compound of formula (I), 25 or, in those embodiments in which AF267B or AF150O(S) may be employed, may be present in the unit dosage in an amount in the range of about 0.5 to about 100 mg. In an embodiment of the invention the compound is present in an amount of about 5 to about 100 mg. In an embodiment of the invention the compound is present in an amount of about 10 to about 50 mg. These amounts may represent a single dose or the total of 2-4 individual doses for administration from 2 to 4 times per day. In an 30 embodiment of the invention, the pharmaceutical composition is in sustained release form. Certain of the compounds in some embodiments of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms, including hydrated forms, are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. 35 The compounds AF150(S) and AF267B have been described in U.S. 5852029. In some embodiments of the invention, the compounds have antioxidant activity. Such antioxidant activity may be the result of such compounds being N-oxides, such as AF600, or it may be the result of such compounds having an anti-oxidant moiety linked at the 4-position nitrogen.
WO 03/092580 PCT/IL03/00357 23 The skilled artisan will appreciate that many factors influence the selection of any compound for application in clinical therapy, e.g., effectiveness for the intended purpose, safety, possible side-effects and therapeutic index. The skilled artisan will thus appreciate how to interpret the expression "pharmaceutically acceptable quaternary compounds" which are structurally derived from the inventive 5 compounds having a tertiary nitrogen atom, as this expression is used in the present specification and claims. The compounds used in embodiments of the present invention can be prepared and administered in a wide variety of oral and parenteral dosage forms. Thus, the compounds used can be administered by injection, that is, intravenously, intramuscularly, intracutaneously, subcutaneously, intraduodenally, 10 or intraperitoneally. Also, the compounds can be administered by inhalation, for example, intranasally. Additionally, the compounds can be administered transdermally. It will be appreciated by those skilled in the art that the following dosage forms may comprise as the active component, either a compound of Formula (I) or a corresponding pharmaceutically acceptable salt of a compound of Formula (I), in accordance with embodiments of the invention optionally with AF267B or AF 150(S) present as well. 15 For preparing pharmaceutical compositions from compounds of formula (I), optionally also including AF267B or AF150(S), pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. A solid carrier can be one or more substances which may also act as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material. 20 In powders, the carrier is a finely divided solid which is in a mixture with the finely divided active component. In tablets, the active component or components is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired. In an embodiment of the invention, the powders and tablets contain from five or ten to about seventy percent of the active compound. Suitable carriers include magnesium carbonate, magnesium 25 stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like. The term "preparation" is intended to include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component with or without other carriers, is surrounded by a carrier, which is thus in association with it. Similarly, cachets and lozenges are included. Tablets, 30 powders, capsules, pills, cachets, and lozenges can be used as solid dosage forms suitable for oral administration. For preparing suppositories, a low melting wax, such as a mixture of fatty acid glycerides or cocoa butter, is first melted and the active component is dispersed homogeneously therein, as by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, 35 and thereby to solidify. Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water propylene glycol solutions. For parenteral injection liquid preparations can be formulated in solution in aqueous polyethylene glycol solution.
WO 03/092580 PCT/IL03/00357 24 Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizing and thickening agents as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, 5 sodium carboxymethylcellulose, and other well-known suspending agents. Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration. Such liquid forms include solutions, suspensions, and emulsions. These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the 10 like. In an embodiment of the invention the pharmaceutical preparation is in unit dosage form. In such form the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Also, 15 the unit dosage form can be a capsule, table, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form. The quantity of active component in a unit dose preparation may be varied or adjusted as recited above, according to the particular application and the potency of the active component. The composition can, if desired, also contain other compatible therapeutic agents. 20 In therapeutic use, the compounds utilized in accordance with embodiments of this invention may be administered at the initial dosage of about 0.01 mg to about 100 mg/kg daily. In an embodiment of the invention, a daily dose range of about 0.01 mg to about 10 mg/kg is used. In another embodiment of the invention, a daily dose range of 10 to 50 mg/kg is used. The dosages, however, may be varied depending upon the requirements of the patient, the severity of the condition being treated, and the 25 compound or compounds being employed. Determination of the proper dosage for a particular situation is within the skill of the art. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day, if desired. 30 The methods used for preparing compounds of the invention include methods which are essentially known to organic chemists for the formation of the five-membered rings, ring-substitution, changing the degree of ring saturation/unsaturation, interconvertion of salts and bases, quaternary salt formation, and so forth. In these synthetic methods, the starting materials may contain a chiral center and, when a racemic starting material is employed, the resulting product is a mixture of R, S 35 enantiomers. Alternatively, a chiral isomer of the starting material may be employed and, if the reaction protocol employed does not racemize this starting material, a chiral product is obtained. Such reaction protocols may involve inversion of the chiral center during synthesis. Select compounds of formula (I) are capable of existing in a number of stereoisomeric forms including geometric isomers such as E and Z (in the nitrones) and enantiomers. The invention includes each of these stereoisomeric WO 03/092580 PCT/IL03/00357 25 forms, and to mixtures thereof (including racemates). The different stereoisomeric forms may be separated one from the other by the usual methods, or any given isomer may be obtained by stereospecific or asymmetric synthesis. It will be appreciated, therefore, that while exemplary methods of preparing certain compounds of the invention will be described, other methods may also be used to 5 prepare the compounds, as will be known by skilled person. When the five-membered ring is thiazolidine-3'-one ring, for example, the compounds may be prepared by forming this ring by reacting the corresponding N-heterocyclic ketone with 2-mercapto carboxylic acid [RxCH(SH)CO 2 H] and ammonia, and the 4-N atom in the product may be substituted in known manner. These reactions may be illustrated as follows in Scheme 1, where the N-heterocyclic 10 ketone is exemplarily 1-methylpiperidine-4-one. R S
H
3 C-N O + RIqHCO 2 H NH 3
H
3 C-N SH O (A)
R
1 S (A) + R 5 -(leaving group) > H 3 C-N O 1 0 R (B) [(A) e.g. AF267: R =Et, R 5 =H;, AF277.: R'=H R'=H); (B) e.g. AF700: Ri=Et, R 5 = 4-fluorobenzenesulfonyl; AF703: R' =Et, R 5 = 2-(1H-indol-3-yl)-ethyl] SCHEME 1 15 The leaving group in "R-(leaving group)" may be e.g. bromide or chloride. This substitution reaction to obtain structure (B) may be conducted under essentially known condition, e.g. by reacting of (A) in presence of an alkali such as lithium di-isopropylamine (LDA) and using a solvent such as tetrahydrofuran (THF). 20 Compound structure (A) or compound structure (B) can be separated into two enantiomers by chiral HPLC, e.g. chiral preparative liquid chromatogrph (LC) separation of AF267 (R'=Et, R=H) gave AF267A [R=(R)Et, R 5= H] and the active enantiomer AF267B [R'=(S)Et, R=H]. A large scale cost-effective method was developed and a maximum 50% yield of each enantiomer can be obtained (yield >97% of each enantiomer, with ee >99% and HPLC purity > 99%). 25 The method can be further expanded for an even more practical separation for those skilled in the art using a Simulated Moving-Bed (SMB) technology for chiral separation as defined by Mazzoti et al (Proceedings of the Chiral Europe 96 Symp, Spring Innovations, Stockport UK, p 103, 1996). AF267B can also be produced by racemization of the AF267A by chemical means, e.g. base-catalysis, or by enzyme-catalyzation followed by chiral HPLC or SMB separation.
WO 03/092580 PCT/IL03/00357 26 Structure (B) may be prepared by reacting the corresponding N-heterocyclic ketone with 2-mercapto acid [RICH(SH)CO 2 H] and primary amine. This reaction may be illustrated as follows in Scheme 2, where the N-heterocyclic ketone is exemplarily 1-methylpiperidine-4-one: R S
H
3 C-N O + R1qHC 2 H / R6NH2 > H 3 C-N SH C NI
R
6 O (B) 5 (e.g. AF282: RI=H, RS=benzyl; AF286: R'=H, R'=2,4-dimethoxybenzyl; AF288: RI=Et, R'=2,4-dimethoxybenzyl; AF285: R 1 '=Et, R'=benzyl) SCHEME 2 Structure (B) may also be obtained by reacting (A) under conditions to obtain amide bond as 10 described below in Scheme 3, where the reacting acid is exemplarily 3-indolpropionic acid: R1 R /S S H3C-N + R 5 -(leaving group) -- H 3
C
N DN H O Rs O R (A) (B) e.g. AF267 Ri=Et R 5 -(leaving group)= AF704 RI=Et 3-indolpropionic acid R5=3-1H-indol-3-yl-propionyl SCHEME 3 The R' group in structure (A) or in structure (B) may be obtained by reaction of the 15 2-unsubstituted compound (A) or (B) with alkyl halide or alkyl aldehyde under standard conditions to effect substitution in the 2-position. These reactions may be illustrated as follows in Scheme 4:
R
1 S S
H
3C -N - H 3 C-N IO ID N N 15R 15 0 RO R AF277 R 5 =H e.g. AF298 RI=CH(OH)CH3,
R
5 -H AF285 Ri=Et, R 5 =benzyl SCHEME 4 20 This method can be applied to 14C-labeling of AF267. The introduction of the ethyl moiety by alkylation of the readily available N-protected AF277 (AF287) with 14C-labeled ethyl bromide, WO 03/092580 PCT/IL03/00357 27 followed by removal of the protecting group yield 1 4 C-labeling of AF267. The synthetic pathway, which may be used analogously to prepare AF267 enriched with 13C, is described below in Scheme 5: ** S N iH S S 0 S NH N N N 'N CN N I I I I
OH
3
OH
3 - OH 3 O H 3 AF277 AF287 [ 1 4 C]-AF267 * * 00 i *.. O ---- q . O S NH S N, H ON-H i. CH20, ... N N I I ii. LDA, THF-DMPU, CH 3
CH
3
CH
3
*CH
2 Br
[
14 C]-AF267A iii. Silica-gel [14C]AF267B iv.Chiral HPLC 5 SCHEME 5 The 1-methyl group in structure (A) and in structure (B) may be removed by reaction with min-chloroperbenzoic acid /FeCl 2 or with demethylating agent such as phenylchloroformate as shown in Scheme 6: R' R S demethylating agent S
H
3 C-N > H-N N N 5 0 15 0 R R 10 (C) e.g. AF504 R'=Et, R=H; AF292=[(S)-AF504] SCHEME 6 AF504 or AF292 may also be prepared by reacting the corresponding N-heterocyclic ketone with 15 2-mercaptocarboxylic acid [R'CH(SH)CO 2 H] and primary amine. This reaction may be illustrated as follows in Scheme 7, where the N-heterocyclic ketone is N-BOC-piperidine-4-one (BOC= tert-Butoxycarbonyl): WO 03/092580 PCT/IL03/00357 28 0 0 0 NH s NH Chiral HPLC
NH
3 TFA separation + EtjHCOzH -- > N SH N H O O O O N-Boc-4-piperidone AF504 0 0 INH S NH S NH + N N I I H H AF291 AF292 SCHEME 7 Stereospecific synthesis of structure (A) or (B) may also be obtained by reacting the corresponding 5 N-heterocyclic ketone with the appropriate 2-mercaptocarboxylic acid, for example: when 1-methylpiperidine-4-one is reacted with (S)-2-mercaptobutyric acid and NH 3 , AF267B is obtained, as shown in Scheme 8 [the (S) configuration is based on the x-ray crystallography of AF267B]: H 00 H S NH H S,, CO 2 H
NH
3 + N N
CH
3
CH
3 (S)-2-mercaptobutyric AF267B acid SCHEME 8 10 (S)-2-Mercaptobutyric acid is commercially available or is prepared from (R)-bromobutyric acid. This reaction may be illustrated as follows in Scheme 9: WO 03/092580 PCT/IL03/00357 29 H H H0 2 C,, NH 2 a H0 2 C,, Br a R-(+)-2-aminobutyric R-2-bromobutyric acid acid H H PhCOS,, CO 2 H HS,,
C
0 2 H c S-2-benzoylthiobutyric S-2-mercaptobutyric acid acid (a)NaNO 3 ,KBr,HBr (b)PhCO-SCs* (c)NH 4 OH SCHEME 9 When compound (A) is reacted with oxidizing agent such as hydrogen peroxide or 5 m-chloroperbenzoic acid, structure (D) or structure (E) is obtained as shown in Scheme 10: R 0 R II S S
H
3 C- N H 3 C-N /I1 O I I O - KN N 0 1 0 I 0 H H (D) (E) [e.g. AF299 R =(S)-Et] [e.g. AF300 R =(S)-Et] SCHEME 10 10 The thio analog of structure (A) or (B) may in general be obtained by reacting the corresponding thiazolidinone ring in structure (A) with Lawesson's reagent, for example as shown in Scheme 11: R (A) + Lawesson's Reagent - HC-N (AF267) (F) 5 S () R e.g. AF510, R = Et, R 5 = H SCHEME 11 15 Bivalent compounds containing essentially two ligands within the same molecule may be obtained by reacting the corresponding compound structure (A) with a spacer under the same conditions to obtained compound structure (B) as described earlier. The spacer-(leaving group) is WO 03/092580 PCT/IL03/00357 30 exemplarily alkaneldihalide, alkanediol, alkanediacid, poly(ethylene glycol). This reaction may be illustrated as follows in Scheme 12: R 0 R 0 0R S N H + (leaving ) " leaving S N,_ (sp acer) N S group )--(spacer)-( group ) SN N 'N)
CH
3
CH
3 (G) CH 3 e.g. spacer = (CH2) n , (CH 2
CH
2 0) n , n = 1-12 leaving group = Br, C(0)CI, C(O)OH SCHEME 12 5 N-Phosphonooxymethyl prodrugs were reported in Krise et al, J Med. Chem. 42: 3094-3100 (1999). Such moieties of N-phosphonooxymethyl can be used also for synthesis of prodrug (H) for improving the water solubility of tertiary amine-containing compounds (B) as shown below. The tertiary amine in compound structure (B) undergoes a nucleophilic substitution reaction with 10 di-tert-butyl chloromethyl phosphate which results in the formation of the quaternary ammonium phosphate protected prodrug. The free acid form of the prodrug is obtained after removal of the tertiary butyl groups as shown in Scheme 13: R O R 0 R sRO0 S NR + /^ S NR + Cl OPOtBuS NR Ot-Bu Cl- -OOCF 3 N N + CH
H
3 C HC O-P-O-t-Bu O-P-OH Ot-Bu OH (H) SCHEME 13 15 The nitrones, compounds of type (I) and (J), can be prepared by reacting the spiro-ketone with alkyl hydroxyl amine or aryl hydroxyl amine. These reactions may be illustrated as follows in Schemes 14A and 14B: R R 1 A A H3C-N + RNHOH H 3 C-N 0 N-O 20 (e.g. AF600: A4=0, R'=Me, RI=Me; AF601: A=0, R'=Et, R'=Me; AF602: A=O, R'=Ph, R5=Me; AF604: A=O, R' =Me, R 5 =benzvl; AF605: A=0, RI =Me, R= iPr) WO 03/092580 PCT/IL03/00357 31 SCHEME 14A R' R' A A A + R 1 NHOH 6 0 N (j) 15 (J) R 5 (e.g. AF603 A=0O, R' =Me, R'=Me) 5 SCHEME 14B Alternatively, structure (I) can be prepared by reacting a five-membered ring carbonyl with alkyl hydroxyl amine or aryl hydroxyl amine. The resulting nitrone is cyclized to form the spiro structure.
CH
3 H3C-N The compound of formula N designated AF150(S), is described in 10 U.S. patent 5,407,938. However, the synthesis of this compound has now been improved. The improved synthesis is described in the following scheme 15:
CH
2
NH
2 CHzNHCOCH 3
CH
2
NHCSCH
3 81% L 83% 79% N (CH 3
CO)
2 0 Lawesson's reagent N NaBH 4 (A) CH 3 1 CH CHI CH 3 CH3 4-picolylamine
CH
3 CHzNHCSCH, N S 75% PPA N N I I
CH
3 CH 3 AF150(S) SCHEME 15 15 AF150(S) was obtained by reaction of 4-picolylamine with acetic anhydride/methyl iodide, followed by reaction with Lawesson's Reagent. The obtained thiopyridinium iodide was reduced to give thioacetylamnino-tetrahydropyridine which was cyclized to form AF150(S). When prepared as a free base, AF150(S) is a colorless liquid. The free base may be stored cold (-20-0 0 C) as a bulk material in dark storage under dry vacuum. AF150(S) may also be obtained as a 20 salt. In an embodiment of the invention, citric acid is used to obtain a stable salt that can be used eventually in a large scale production. A white crystalline citric acid salt of AF150(S) was prepared by WO 03/092580 PCT/IL03/00357 32 mixing AF150(S) free base with citric acid in 2-propanol and tetrahydrofuran solution. In comparison to the free base, the salt: (a) lacks color development even at high temperature (accelerated stability test), and (b) shows high stability if the bulk is kept under anhydrous conditions even at high temperature (accelerated stability test). 5 In another embodiment of the invention, AF150(S) is provided in pharmaceutical acceptable paraffin oil. The stability of 10% w/w AF150(S) in paraffin oil was examined at 40 0 C, under air or nitrogen atmosphere and in the presence or absence of tocopherol. No degradation products above 0.1% were detected. A slight yellow color was observed in samples without tocopherol but color was not developed in samples containing 0.5% w/w tocopherol in AF 150O(S). 10 It may noted that the N-methyl group in AF150(S) may be removed by reaction with mn-chloroperbenzoic acid /FeC12 as shown in Scheme 16: COH 3 CH 3 HC-N NH-N NN AF150(S) AF400 SCHEME 16 15 When AF150(S) is reacted with oxidizing agent such as mn-chloroperbenzoic acid, AF406 is obtained.
CH
3 0- N H C -N+ O AF406 Cold simulation to 1 4 C-labeling of AF150(S), AF402, was obtained by using d 3 -iodomethane instead iodomethane in the first step of the synthesis, according to the scheme of the synthesis of 20 AF150(S). The synthesis of AF402 is described in Scheme 17 below:
CH
2
NH
2 (CH3CO)20 CH 2
NHCOCH
3
CHNHCSCH
3
CD
3 1 +, 10 + N N N I I (A)
CD
3
CD
3 4-picolylamine
CH
3
CHNHCSCH
3 - N S N N I CDa
CD
3 AF402 SCHEME 17 WO 03/092580 PCT/IL03/00357 33 It is to be understood that whereas in the foregoing description, the illustrative compounds of the invention have shown piperidine, and quinuclidine rings, other any nitrogen-containing heterocyclic rings suitable for spiro-configuration with the depicted spiro five-membered ring may be substituted 5 therefore. Such compounds may be made by using the corresponding ketone, in analogy to the use of 3- or 4-piperidone to obtain compounds shown above. A similar remark applies to the practical EXAMPLES, which are merely illustrative and not limitative. The invention will now be illustrated by the following non-limiting EXAMPLES. 10 EXAMPLE 1 Synthesis of (S)-2-Ethyl-8-methyl-1-thia-4,8-diaza-spirof4.57decan-3-one 0 E" E NH N 1
CH
3 AF267 B Step 1: Synthesis of 2-mercaptobutyrfic acid. 15 2-Bromobutyric acid (3.2 kg, 19.16 mol) was introduced in a cooled (ice-water bath) flask. It was stirred and aqueous potassium hydroxide (18.2 mol) was added gradually (0.5 h) while the temperature was maintained at 30-40'C. Cooling was stopped and potassium O-ethyldithiocarbonate (3.48 kg, 21.7 mol) was added in portions so that the temperature did not exceed 50'C (0.5 h). The reaction mixture was stirred at 50'C for 1 hour, then cooled to 15-200C (ice-water bath). Ethylenediamine (2.6 1, - 39 20 mol) was added during a period of 20 min while the temperature was maintained at 45-55 0 C (external cooling). The resulting suspension was stirred at 500C for two hours, cooled to 20'C, filtered and the solid was washed with 2 x 1.5 liter warm water (40-50 0 C). The aqueous filtrate and washings were combined, cooled below 200C (ice-water bath) and aqueous sulfuric acid [5.2 1, 50%(w/w)] was added slowly while keeping the temperature at 45-55 0 C (0.5 hour, to pH=2). The solution was cooled to 30 0 C 25 and transferred to a 25 liter container equipped with a mechanical stirrer. Methyl-t-butyl ether (MTBE, 3 1) was added and the mixture stirred and left overnight at room temperature. The upper oily phase was separated and the lower aqueous phase was filtered under reduced pressure to remove a precipitate which was formed. The aqueous phase was extracted with MTBE, the extracts were combined and the MTBE was removed. The oily residue was dissolved in cyclohexane (5 1) and kept in a refrigerator 30 overnight. A lower phase was formed. It was separated and extracted with cyclohexane. The cyclohexane extracts were combined with the upper phase, cyclohexane was removed and the WO 03/092580 PCT/IL03/00357 34 2-mercaptobutyric (crude) acid was dried in vacuum (54 0 C/2 mmHg) to yield 2-mercaptobutyric acid (2.18 kg, 18.16 mol). Step 2: Synthesis of AF267 (racemate). 5 2-Mercaptobutyric acid (705 g, 5.88 moles) and a mixture of cyclohexane/tert-butyl alcohol (1;3.5(w/w), 4.3 1) were introduced into a flask. The solution obtained was stirred and heated to 40-60 0 C. Gaseous ammonia was bubbled through the solution till all or most of the 2-mercaptobutyric acid was converted to its ammonium salt. The bubbling of the ammonia was stopped, the reaction mixture was heated to reflux and a solution of 1-Methyl-4-piperidone (496 g, 4.39 mol) in a 10 cyclohexane/tert-butyl alcohol mixture (500 ml) was added. After 1 hr, the solution become clear and the bubbling of ammonia was renewed and after 13 hrs (addition of piperidone and reaction time afterwards) the reaction mixture was cooled and left overnight at room temperature. Hydrochloric acid solution prepared by diluting aqueous concentrated acid (one volume) with water (two volumes) (960 ml) was added and the mixture stirred for 1 h. The solution obtained (pH-2-3) was cooled to 25 0 C and 15 the lower aqueous phase was separated and made basic with aqueous potassium hydroxide (pH-8.5-9) then left overnight at room temperature. The product which precipitated was filtered and washed with cold water (100 ml) to give wet powder. The filtrate was basified to pH 9 and left overnight at 5 oC, filtered and washed with 50 ml cold water to give 72 g powder.The same procedure was repeated to synthesize a second batch of AF267 (multiplied by a factor of 1.2). The corps were combined to give 20 1.6 kg of wet product. The crude combined product was dissolved in 4.5 liter of hot water (95°C), filtered and the clear solution was left at room temperature for 10 hrs, filtered and dried for 24 hrs (50 'C, 1 mmHg) to give AF267 (1.048 kg, 50.7% yield). The filtrate was concentrated, cooled overnight at 5 0 C, filtered, washed (100ml cold water) and dried to give AF267 (215 g, 10.4% yield). Total AF267 yield: 61%. mp. 142-144 'C; 'H NMR (CDC1 3 ) 3 1.02 (t,j= 7.3Hz, CH 3
CH
2 ), 1.7-1.8 (min, CH 3 CHH), 25 1.96-2.07 (m), 2.30 (s, NCH 3 ), 2.3-2.36 (min, 2H), 2.6-2.7 (bs, 2H), 3.80 (dd, j = 8.7, 3.9Hz, 1H, SCH) ppm. MS rm/e 214(M+), 181(M-SH); Anal. (C 1 0
H
1 8
N
2 0S) called. C 56.04, H 8.47, N 13.07, S 14.96; found C 55.92, H 8.44, N 13.23, S 14.81. Step 3: Chiral separation of AF267B and AF267A 30 Prochomrn LC 110 High Performance Preparative Liquid Chromatograph Column: CHIRALPAK9ASV (lot number JG 001) Pump flow rate: 500 ml/min Pressure: 12.7 bar Column Temp 26 0 C 35 Moblie phase: Acetonitrile/EtOH 85:15 Concentration: 37 gr/1 UV Detection: 240/230 umn Following elution the eluent was evaporated to dryness. First eluting enantiomer (AF267A): ee: 99.7 (687.1 gr; purity 99.3% ; Yield: 97%) WO 03/092580 PCT/IL03/00357 35 Second eluting enantiomer (AF267B): ee: 99.8 (694.9 gr; 99.9% ; Yield: 98%). Residual solvent (e.g. acetonitrile) was removed by further addition of ethanol and evaporation to dryness. By analogous syntheses, AF292 and other related compounds may be prepared. 5 EXAMPLE 2: X- Ray Single crystal structure analysis of (S)- 2-Ethyl-8-methivl-1-thia-4,8-diaza-spiro [4. 57decan-3-one (AF267B) 10 Crystal data: Co 10 Hi 9 NOS+ H 2 0 (monohydrate), transparent, light yellow, prisms, crystal size 0.2x0.2x0.4 mm 3 ; crystal system, orthorhombic, space group: P212121 (No 16) a=10.394 (10) (ca=90'), b=20.133 (2) (f3=90 0 ), c=5.856 (4) (y=90 0 ), A, from 25 reflection, T=110K, Volume=1224.2 (9) A 3 , Z=4, Fw=202.32, Calculated density, Dc=1.092Mg/m 3 , Absorption coefficient, g =0.232mm " 1 . Data collection and treatment: Rigaku AFC5R four-circle diffractometer, MoKc, graphite 15 monochromator (X=0.71073 A), 11461 reflections collected, Theta range for data collection: 2.820 5 0 5 27.53°; Index ranges: -13 < h <13, -26 < k < 26, 0 < 1 <7, o) scan method, scan width=1.2o, scan speed 2 0 /min, typical half-height peak width=0.45 0 , 3 standards collected 62 times each, with a 3% change of intensity; Reflections collected: 6272 measurements, 2833 independent reflections [R (int)=0.0604, Bijvoet reflections kept separated]. 20 Solution and refinement: structure solved by direct methods (SHELXS-97). Full-matrix least-squares refinement based on F 2 (SHELXL-97). Idealized hydrogens were placed and refined in a riding mode, water hydrogens found from the difference Fourier map, 148 parameters; final R indices: R 1 =0.0671 (based on F 2 ) for data with I >2 sigma(I) wR 2 =0.1484 and RI=0.0747 wR 2 =0.1553 for all data, goodness-of-fit on F 2 =1.13, largest electron density =0.745ef 3 .. around S atom. 25 Absolute configuration: The absolute configuration of the molecule was determined using Flack's parameter approach and the alternative refinement of the enantiomeric twinning component. Both methods show unequivocally that the present coordinates belong to the correct absolute configuration (S enantiomer). The cystals of this compound were prepared from crystallization in toluene/petroleum ether/methanol.
WO 03/092580 PCT/IL03/00357 36 H14a H14b C14 Hl4c H7a NB H9b C7 HBa C6 10b b 9 H9a C 10 W15a H6b 10a C5 015+ H S1 44 H15a C3 C2 H12c H11 2 013 C11 C12 HIlb H12b H12a Structure 1: ORTEP structure of C 10
H
1 9 NOS+ 1H20 EXAMPLE 3: 5 Chiral synthesis of(R>. 2-Ethyl-8-methy-l-thia-4,8-diaza-spiro4.5]decaa-3-one, AF267A H O NH N AF267A The asymmetric synthesis of AF267A was performed in order to model the synthesis of the S enatiomer by using optically active 2-mercaptobutyric acid in the synthesis of AF267. Optically active 2-mercaptobutyric acid was synthesized starting from L-(+)-2-amninobutyric acid (I) which has the S 10 configuration. The amino acid was converted to (S)-2-bromobutyric acid (II) with retention of configuration by treatment with sodium nitrite, potassium bromide and hydrobromic acid. The enantiomeric purity of the obtained bromide was checked by proton NMR measured in the presence of (R)-(+)-N-benzyl-a-methyl-benzylamine and compared to the spectrum of the racemic bromide measured at the same conditions. The presence of only one enantiomer was detected by this method. 15 The bromide was converted to (R)-2-benzoylthiobutyric acid (III) (with inversion of configuration) by treatment with cesium thiobenzoate in DMIF. Debenzoylation of (IIH) was accomplished without racemization by aminolysis (IN ammonium hydroxide at room temperature). The obtained (R)-2-mercaptobutyric acid was purified by distillation and then reacted with ammonium acetate and 1-methyl-4-piperidone in boiling cyclohexane. The crude reaction mixture was WO 03/092580 PCT/IL03/00357 37 analyzed by GC on a chiral column and found to contain AF267A accompanied by a small (2-3%) amount of AF267B. EXAMPLE 4: 5 Chiral synthesis of (S)- 2-Ethyl-8-methyl- -thia-4,8-diaza-spiror4.51decan-3-one AF267B This compound is obtained as in EXAMPLE 3, except that the starting material used is (R)-2-bromobutyric acid. EXAMPLE 5: 10 Synthesis of(S)- 2-Ethyl-I -thia-4,8-diaza-spirof4.5]decan-3-one, AF292 aC3 H 2 _ 0 S NH N I H AF292 To a cooled (ice-salt bath) stirred solution of AF267B (6.lgr, 0.028mol) in dichloromethane (60 ml) was added m-chloroperbenzoic acid (mCPBA) in small portions over a period of 15 min mCPBA (70%, 7.02gr, 0.028mol total). The mixture was stirred for 1 hr and then treated with iron (II) chloride 15 (2.14ml of lM solution in water). Stirring and cooling (-10 'C) were continued for 1 hr and then stirring continued for 2 hrs at room temperature. Ethylene diamine (1.9 ml, 0.285 mol), sodium hydroxide (30.5ml of 2N aqueous solution), and petroleum ether 40-60'C (60 ml) were added. After vigorous shaking the layers were separated, the aqueous layer was extracted with mixture of dichloromethane/petroleum ether 1:1 (600ml) followed by dichloromethane (first 600ml then 300ml). 20 The combined extracts were dried (NakSO 4 ), filtered and the solvents were removed under reduced pressure. Flash chromatography (silica-gel 60, 230-400 mesh, Merck 1.09385, elution with methanol/chloroform/ammonium hydroxide 10: 89:1 v/v) of the residue gave AF292. 1H NMR (CDC13) 5 1.02 (t,j =7Hz, 3H), 1.76 1.92 and 2.07 (3xm, 6H), 2.84 (m, 2H), 3.04 (min, 2H), 3.83 (dd,j =8.8,4Hz, 1H), 7.66 (NH) ppm; 1 3 C NMR (CDCl 3 ) 8 11.50, 27.24, 42.87, 43.95, 44.10, 48.80, 64.22, 175.22 ppm: 25 MS m/e 200(1W). The compound was >99.9% purity by HPLC, GC. The hydrochloride salt of AF292 was formed by addition of HCI (4M in methanol) and recrystallised from methanol-diethyl ether to give a white precipitate that was filtered and dried. 1H NMR (D 2 0) 5 0.78 (t,j=7.3Hz, 3H), 1.58 (min, 1H), 1.76 (min, 1H), 2.05 (min, 4H), 3.07 (in, 2H), 3.30 (inm, 2H), 3.92 (dd,j = 7.9, 4.0Hz, 1H) ppm. 30 WO 03/092580 PCT/IL03/00357 38 EXAMPLE 6: Reactions for preparation of [1 4 C]-2-Ethyl-8-methyl- 1 -thia-4. 8-diaza-spiro F4.51decan-3-one: R and S isomers * 0 * 0 S N- S N. N N I I
[
1 4 C]-AF267B [ 14 C]-AF267A 5 In a septum-capped dried flask (10 ml) equipped with a magnetic bar and nitrogen inlet, a solution of diisopropylamine (0.078 ml, 0.56 mmol) in dry THF (1.4ml) is introduced by a syringe and cooled to 0 0 C. n-BuLi (0.9 M in hexane, 0.62 ml, 0.56 mmol) is added, the reaction mixture is stirred at 00C for 20 min and then cooled to -78 0 C. A solution of AF287 (0.1375 gr, 0.51 mmol) in dry THF (0.4 ml) and dry N,N'-dimethyl-N,N'-propylene urea (DMPU) (0.6ml) is added dropwise (30 min) and 10 the reaction mixture is stirred for 20 min at -780C. Ethyl bromide (0.043ml, 0.56mmol) labeled at the 1-carbon with 140 is added in one portion, the temperature is allowed to rise to room temperature and the reaction mixture is stirred for an additional 4h. The solvents are removed under reduced pressure, first using a water pump at 25 0 C for 20 min and then an oil pump (-4mm Hg) at -60'C for ~30 min (using a needle which introduced a stream of air to remove the solvent faster). Flash chromatography of 15 the residue gives racemic AF267 (95mg, ~86% yield). Preparative chiral HPLC may be used to separate the enantiomers. By following the above procedure using ethyl bromide which was not labelled, preparative HiPLC of 60mg of the racemate obtained after flash chromatography afforded non-isotopically labeled AF267B (17.6mg). 20 EXAMPLE 7: Synthesis of (S)-2-Ethyl-8-mnethvl-8-oxy-1-thia-4.8-diaza-spirof.57decan-3-one, AF299
CHCH
2 , ._ 0 S NH -0 1
CH
3 AF299 A solution of mCPBA (70%, 2.
6 2gr, 10.64mmol) in dichloromethane (40ml) was added slowly (0.5 hr) to a cold (0 0C) and stirred solution of AF267B (2.07gr, 9.67mmol) in dichloromethane (40ml). 25 The cooling bath was removed and the reaction mixture was stirred at room temperature for 2 hrs and then the solvent was removed under reduced pressure. Flash chromatography (methanol/chloroform/ ammonium hydroxide 10:89:1 v/v) of the residue and precipitation of the product as a solid from methanol-acetonitrile gave the N-oxide, AF299; 1 H NMR (CDC1 3 ) 8 0.99 (tj = 7.3 Hz, CH 3
CH
2 ), 1.74 WO 03/092580 PCT/IL03/00357 39 and 2.09 (2m, CH 3 CH), 1.82 (m, 2H), 3.9 (m, 2H), 3.36 (s, CH 3 N+), 3.33-3.45 (m, 4H), 3.78 (br NH), 3.83 (dd, j= 3.74, 8.84 Hz, CH 3
CH
2 CR) ppm; MS m/e 230 (M). EXAMPLE 8: 5 Synthesis of (S)-2-Ethyl-8-mnethyl-1-oxo-l4-thia-4,8-diaza-spirof4.57decan-3-one. AF300 CHzCH 0O NH N
CH
3 AF300 A solution of AF267B (1.72gr, 0.008mol) in water (2.5ml) was cooled (ice-water bath) and trifluoroacetic acid (3.5 ml) was added. To the cold stirred obtained mixture was added hydrogen peroxide (30%, 0.57ml, 0.008mol), the cooling bath was removed and the reaction mixture was stirred 10 at room temperature over night. Sodium sulfite was added and the pH of the solution was adjusted to 9 with a saturated solution of sodium carbonate. The aqueous phase was extracted with dichloromethan (2x00ml) and then with ethyl acetate (lxS0ml). The organic extracts were combined, dried (MgSO 4 ) and the solvent was evaporated. Flash chromatography (silica-gel 60, 230-400 mesh, Merck 1.09385, elution with methanol/chloroform/ammonium hydroxide 10:89:1 v/v) gave AF300. 1 H NMR (CDC1 3 ) 5 15 1.19 (t,j= 7.3Hz, 3H, CH 3 ), 1.85-2.0 (m, 5H), 2.17 (m, 1H11), 2.27 (m, 1H), 2.34 (s, 3H, NCH 3 ), 2.6 (m, 2H), 3.30 (dd,j = 3.56, 11.13 Hz, CH 3
CH
2 CH), 6.37 (br NH) ppm; MS (EI) m/e 230 (M). EXAMPLE 9: Synthesis of 2-(I-Hydroxv-ethyl)-8-methyl-1 -thia-4,8-diaza-spiro f4.51decan-3-one (AF298) HO - 0 S NH N 20 A298 To a cold (0'C) solution of diisopropylamine (0.28ml, 0.002mol) in dry THF (12ml) under argon atmosphere was added a solution of n-butyllithium (1.4M in hexane, 1.4ml, 0.002mol), the mixture was stirred for 20 min and then cooled to -78 0 C. A solution of AF287 (0.41g, 0.0015mol) in THF (3ml) was added dropwise (10min) and the resulting mixture was stirred at -78 0 C for additional 10min. 25 Acetaldehyde (0.85ml, 0.015mol) was added in one portion and after ten min at -780C acetic acid (0.11ml, 0.002mol) was added in one portion and the temperature was allowed to raise to room temperature. The reaction mixture was added to chloroform (200ml) and the organic phase was washed with water (2x20ml), separated and dried. The solvent was evaporated and flash chromatography WO 03/092580 PCT/IL03/00357 40 (silica, CHC1 3 /MeOHNH4IOH 80/20/1) of the residue gave AF298 (0.132g). 'H-NMR (CDC1 3 ) 5 1.24 (d,j=6.05Hz, 3H, CH 3 ), 1.7-2.2 (min, CH2), 2.31 (s, 3H, NCH 3 ), 2.60-2.80 (min, CH 2 ), 3.71 (d,j = 9.41Hz, 1H, SCH), 3.94-4.04 (min, 1H, CHO), 4.75-4.9 (br OH), 7.1-7.2 (br NH) ppm. MS m/e 230 M(+). 5 EXAMPLE 10: Synthesis of (S)-2-Ethyl- 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza- spiroF4.51decan 3-one. AF700 0 N I AF700 Into a cold (0 'C, ice-water bath) solution of lithium hexamethyldisilazane (8ml, lM in THF) was 10 added AF267B (1.5g, 0.007mol) in small portions over a period of 15 min under argon atmosphere. The cooling bath was removed and the reaction mixture was stirred at room temperature for 40 min. 4-fluorobenzylsulfonyl chloride (1.37g, 0.007mol) was added (during the addition the temperature was kept below 20 oC, cooling bath) and the reaction mixture was left at room temperature under argon atmosphere overnight. Dichloromethane (100 ml) was added. The reaction mixture was washed with 15 water (20 ml) the organic phase was separated, dried (MgSO 4 ) and evaporated. Flash chromatography (silica, ethyl acetate/methanol/aqueous ammonia 10/2/0.1) gave AF700 (0.46gr, 0.0015mol). 1 H-NMR (CDC1 3 , 300 MHz) 8 0.94(t, j = 7.14 Hz, 3H, CH 3 ), 1.64-1.77(m, 3H, CHH+ CH2), 1.88-2.00(m, 3H, CHH + CH2), 2.15-2.29(br, 2H, CH 2 ), 2.29(s, 3H, NCH 3 ), 2.51-2.60(br, 2H, CH2), 4.24(dd, j = 7.99,3.72 Hz, 1H, SCH), 7.21(app. t,j = 8.53 Hz, 2H, Ar), 8.07-8.12(m, 2H, Ar). 20 EXAMPLE 11: Synthesis of 8-Methyl- 4-pyrrolidin- 1-vlmethyl- 1-thia- 4,8-diaza- spiro[4.51 decan- 3-one. AF287 0 N AF287 A solution of 8-Methyl-1-thia-4,8-diaza-spiro[4.5]decan-3-one (AF277 5.13g, 0.0275 mol), 25 formaldehyde (37% solution, 2.75m1), and pyrrolidine (2.3m1, 0.0275mole) in ethanol (2.3ml) was refluxed for 4h, then left at room temperature over night. Toluene (10ml) was added and the solvent was evaporated. Boiling pentane (100ml) was added to the residue and the solution was decanted. The trituration with hot pentane was repeated four times, the pentane layers were combined, cooled to 0OC and the precipitate was collected and identified as AF287 (4.6gr, 63% yield). 'H-NMR (CDC13) 8 WO 03/092580 PCT/IL03/00357 41 1.69-1.77(m, 5H), 2.20-2.29(s and min, 5H, CH 3 and CH 2 ), 2.43-2.58(m, 6H), 2.82-2.86(m, 2H), 3.50 (s, 2H, SCH 2 ), 4.14(s, 2H, NCH 2 N)ppm. MS (EI) m/e 269(M+); 198; 84; 70. EXAMPLE 12: 5 Synthesis of 2-Ethyl- 4-(3-1H-indol-3-vl-propionvl)- 8-methyl- 1-thia- 4.8-diaza- spiror4.5] decan-3-one. AF704 0 O N n _H N I AF704 A solution of AF267 (1.2gr, 0.0056mo1), 3-indolpropionic acid (1.37gr, 0.0072mo1), dicyclohexylcarbodiimide (DCC) (1.57gr, 0.0076mol) and 4-dimethylaminopyridine (DMAP) (0.93gr, 10 0.0076mol) in dichloromethane (120ml) was stirred at room temperature for 3 days. The reaction mixture was washed with water (2x40ml), the organic phase was dried and evaporated. Flash chromatography (silica, CHCl 3 /MeOH/NH 4 OH 90/10/1) gave the title compound which was triturated in acetone. The solution was filtered to remove the impurities, then the acetone was evaporated and the obtained thick oil was triturated in ether. The obtained solid (AF704), 400mg, was filtered and dried. 15 Mp. 122.5-124.5 °C; 'H-NMR (CDCI 3 ) 8 1.01 (t,j = 7.4Hz, 3H, CH 3
CH
2 ), 1.50 (m, 1H), 1.59 (min, 1H), 1.61-1.73 (min, 1H, CH 3 CHH), 2.1-2.15 (min, 1H, CH 3 CHH), 2.18 (dt, j= 12.4, 2.4Hz, 1H), 2.29 (s, 3H,
NCH
3 ), 2.33 (in, 1H), 2.83 (min, 2H), 2.99 (dt, j = 12.55, 4.39 Hz, 1H11), 3.01 (t,j = 7.42Hz, 2H11, CH2), 3.16 (dt, j = 12.7, 4.39Hz, 1H), 3.28 (min, 2H11), 3.67 (dd, j = 8.9, 4.18Hz, 1H, SCIHI), 7.04 (br s, C=CH), 7.11 (t,j = 7.8Hz, ArH), 7.18 (t,j = 7.1Hz, ArH), 7.34 (d,j = 8.08Hz, ArH), 7.64 (d,j = 7.58Hz, ArH), 20 8.01 (br NH) ppm; MS (EI) m/e 385 M(+), 214, 181, 171,143, 130 (100%). When the starting material is AF267B, the enantiomer AF704B is obtained. EXAMPLE 13: Synthesis of 2-Ethyl- 4-[2-(1H-indol-3-vyl)-ethyll- 8-methyl- 1-thia- 4.8-diaza-spiro [4.51decan- 3-one, 25 AF703 0 S_ N CN) H AF703 In a three-necked flask equipped with a magnetic stirrer and Dean-Stark and dropping funnel a solution of 2-mercaptobutyric acid (7.38gr, 0.065mol) in a mixture of t-butanol/cyclohexane (30/104 WO 03/092580 PCT/IL03/00357 42 gr/gr, 86ml) was heated to 40 'C. Tryptamrnine (10.8gr, 0.068mol) was added in four portions (20min), the reaction mixture was stirred for additional 45min. and then heated to reflux. A solution of 1-methyl-4-piperidone (6.18ml, 0.049mol) in t-butanol/cyclohexane (30/104 gr/gr, 10ml) was added dropwise during 40min and the reflux continued for Shrs (1ml water was collected). Mixture of 5 HCl/water (2:2 v/v) was added until pH 2-3, the aqueous phase was separated, basified to pH 10 with solution of potassium hydroxide and extracted with dichloromethane. The organic phase was separated, dried and the solvent was evaporated. Flash chromatography (silica, CHC1 3 /MeOH/NH 4 OH 90/10/1) of the residue gave the title compound. Recrystallization from boiling hexane gave pure AF703. 'H-NMR (CDC1 3 ) 6 1.04 (t,j= 7.35Hz, 3H, CH 3 ), 1.65-1.68 (min, 2H), 1.71-1.80 (min, 1H, CH 3 CH), 2.13-2.34 (m, LO 5H), 2.31 (s, 3H, NCH), 2.86 (m, 2H), 3.05-3.10 (min, 2H), 3.41-3.49 (min, 1H), 3.51-3.65 (mn, 1H), 3.80 (dd, j = 8.8, 3.9Hz, lIH, SCH), 7.02 (d,j = 2.32Hz, 1H, ArHf), 7.14 (ddd, j = 7.35, 7.35, 1.17Hz, 1H, ArH), 7.20 (ddd, j = 7.45,7.45,1.39Hz, 1H, ArH), 7.35 (d,j = 7.4Hz, 1H, ArH), 7.77 (d,j = 7.42Hz, IlH, ArH), 8.03 (brNH) ppm. 15 EXAMPLE 14: Synthesis of 2,8-Dimethyl-1l-thia-3.8-diaza-spirof4.5]dec-2-ene AF150(S)
CH
3 -N S N AF 50(S) Step 1: Synthesis of 4-(acetaminomethyl)-l-methyl-pyridinium iodide. To a cold (ice-water bath) solution of 4-picolylamine (1070gr, 9.9mol) in methanol (3 1) was 20 added dropwise acetic anhydride (1400gr, 13.7mol). During addition the reaction temperature was kept between 10 and 300C. When the addition of the reagent was complete the reaction mixture was left overnight at room temperature. Iodomethane (800ml, 24.8mol) was added to the reaction mixture which was cooled with water bath, under nitrogen atmosphere. During addition the reaction temperature was kept below 25oC. When the addition was complete, the reaction mixture was protected from light and 25 left at room temperature overnight. The excess iodomethane was evaporated, crystallization was induced, the reaction mixture was cooled (ice bath) and isopropanol (1.5 1) was added. The reaction mixture was left at -30°C overnight, the precipitate was filtered off, washed with isopropanol and dried. 4-(Acetaminomethyl)-l1-methyl-pyridinium iodide (2043gr, 7mol) was obtained as yellow powder (71%yield). 'H-NMR (D 2 0) 8 2.10 (s, CH 3 CO), 4.32 (s, CH 3 N+), 4.63 (s, CH 2 INHCO), 7.89 (d, j = 30 6.6Hz, 2H), 8.67 (d,j = 6.6Hz, 2H) ppm.
WO 03/092580 PCT/IL03/00357 43 Step 2: Synthesis of 1-methvl-4-N-thioacetyaminomnethylpyridinium iodide A stirred solution of 4-(Acetaminomethyl)-1-methyl-pyridinium iodide (1.92kg, 6.5mol) and Lawesson's reagent (1.87kg, 4.6mol) in acetonitrile (6 1) was warmed to 80 0 C for 17hrs. Then the reaction mixture was cooled to room temperature and stirred for an additional 4hrs. The crude 5 thioamide was filtered off and washed with acetonitrile (2.8 1). To the obtained thioamide was added ethyl acetate (10 1) and the suspension was refluxed forlh. At 72-730C a noxious gas was evolved and trapped with NaOH solution. The suspension was cooled to 60 0 C. The thioacetamide was filtered off, washed with ethyl acetate (2 1) and dried (45°C). 2kg of thioamide was obtained. 10 Step 3: Synthesis of 1- methyl-4-N-thioacetylamino-1,2,3,6-tetrahydropyridine. A suspension of 1-methyl-4-N-thioacetylaminomethylpyridinium iodide (2.07kg, 6.7mol) in water (6.2 1) was prepared in 25 1 flask and stirred at room temperature. A solution of sodium borohydride (383gr, 10.1mol) in water (1.2 1) was added dropwise over a period of 3.5 h so that the temperature was maintained below 32 0 C. The reaction mixture was stirred for 2 h at room temperature, 15 ethanol (600ml) was added and stirring was continued for 30 min. Sodium carbonate (780 g) was added and the reaction mixture was stirred overnight. Dichloromethane (4 1) was added, and stirring was continued for 45 min. The solution was filtered in order to remove the solid which was washed with chloroform (1 1) and water (0.5 1). The filtrate was decanted, and the aqueous phase was extracted with chloroform (2.5 1). The organic phases were combined and washed with 10% sodium thiosulfate 20 solution (2.2 1). The aqueous phase was extracted with chloroform (1 1), the organic phases were combined, dried over magnesium sulfate, filtered and concentrated. Acetone (1.5 1) was added, the suspension was stirred for 30 min at room temperature and for 45 min at 0 0 C. The thioacetamide was filtered off, washed with acetone (1.5 1) and dried (50 0 C). 860gr of thioacetamide was obtained. mp. 140'C; 'H NMR (CDCI 3 ) 5 2.21 (m, 2H), 2.36 (s, CH 3 N), 2.56 (s, CH3CS), 2.57 (t, 2H), 2.95 (mn, 2H11), 25 4.23 (d, CH 2 NH), 5.65 (in, CH=C), 7.41 (bs, NH) ppm. MS m/e 184 (M), 151,150, 149, 141, 140, 126, 114, 109 (100%), 109, 96, 94, 82, 70. Step 4: Synthesis of 2.,8-dimethyl-l-thia-3.8-diaza-spiro[4.5]dec-2-ene AF150(S) In three-necked round bottom flask (5 1) equipped with a mechanical stirrer, polyphosphoric acid 30 (2.1kg) was stirred and heated to 100 'C. The thioacetamide (840gr) was added in small portion. At the end of the addition the temperature was raised to 170 'C and this temperature was maintained for 2-3hrs. The hot reaction mixture was slowly poured into a stirred cold aqueous solution of sodium carbonate (25%, 10 1). The basicity was raised by addition of aqueous sodium hydroxide solution (50%, 350ml). The reaction mixture was extracted with chloroform (2x3 1), the organic phases were 35 combined, dried (Na 2 SO4) and evaporated. The residue was dissolved in petroleum ether (3.5 1) to give a solution and small amount of insoluble material. Evaporation of the petroleum ether solution gave crude AF150(S). The described procedure was repeated and the crude AF150(S) from the combined batches was treated with activated carbon and was distilled twice under reduced pressure (0.5mmHg, 61 WO 03/092580 PCT/IL03/00357 44 oC) to give AF150(S) (>1.2Kg, global yield of 40%). 'H NMR (CDC1 3 ) 8 1.8-2.0 (min, 4H), 2.18 (t, 3H,
CH
3 C), 2.28 (s, 3H, CH 3 N), 3.9 (m, 2H, CH 2 ) ppm; IR (C=O) 1636 cm 1 ; MS. M/e 184 (M+). EXAMPLE 15: 5 Synthesis of 2.8-Dimethvl-1-thia-3.8-diaza-spiro[4.51]dec-2-ene 8-oxide AF406 N S + N 1no AF406 A solution of m-chloroperbenzoic acid, mCPBA, (1.40gr, 8.11 mmol) in dichloromethane (3 Oml) was added gradually to a solution of AF150(S) (1.45gr, 7.88mmol) in dichloromethane (10ml). The reaction was stirred at room temperature overnight. Chromatography of the reaction mixture was on a 10 column of natural aluminum oxide (Merck)(metanol:chloroform 1/49) gave AF406 (0.75gr) as a crystalline solid. A sample was crystallized from ethylacetate. A very hygroscopic solid was obtained. mp. 130-132 'C (145-159 'C dec.); 'H NMR (300MHz, CDC13) 5 1.89 (m, 2H), 2.2 (t, j = 1.5Hz,
CH
3 C=N), 2.82 (m, 2H), 3.24 (m, 2H), 3.25 (s, CH 3 N+O-), 3.35 (min, 2H), 4.03 (q,j = 1.5Hz, CH 2 N=C) ppm; IR(CHC13) 2947,1636,1448,1153,931, 664 cmu'; MS(EI) 200 (M), 184,182, 149, 141, 140, 126, 15 110, 109, 108, 96, 82, 70. EXAMPLE 16: Synthesis of 2-Methyl-1-thia-3,8-diaza-spiro[4.5]dec-2-ene, AF400 N S N H AF400 20 A solution of AF406 (2gr, 0.01mol) in chloroform (10ml) was cooled to -10 0 C - -5 0 C in an ice-salt bath. A solution of FeCl 2 (IM, 0.7ml) was added and the two phase reaction mixture was stirred for 4.5h. The color of the reaction mixture changes with time from dark green to dark orange-brown. To the cooled reaction mixture was added cautiously a mixture of petroleum-ether (10ml), ethylene diamine (600mg, 0.01mol) and 2N sodium hydroxide (10ml, 0.02mol). The pH of the water phase was 25 13. The organic was separated and the aqueous phase was extracted with chloroform, acidified with 5N HC1 to pH = 9 and extracted again with chloroform. All the organic fractions were combined, dried on potassium carbonate, filtered and evaporated. Chromatography [silica-gel (RIEDEL DE Haen 31607), WO 03/092580 PCT/IL03/00357 45 CHC1 3 :MeOH:NH40 90/9/1] of the residue gave AF400. mp. 40 oC; 'H NMR (CDC1 3 ) 5 1.74(m, 2H), 1.92 (min, 2H), 2.19 (t,j = 1.8Hz, 3H11), 2.71 (min, 2H11), 3.05 (min, 2H), 3.92 (q,j = 1.8Hz, 2H) ppm; MS m/e 171 (M++l1), 170 (M). 5 EXAMPLE 17: Synthesis of 2-Methyl-8-methyl-d-1-thia-3,8-diaza-spirof4.5]dec-2-ene. AF402 N S N I
CD
3 UU3 AF402 To a stirred cold (ice-water bath) solution of 4-picolyamine (50 g, 0.462mo1) in methanol (200ml) acetic anhydride (75 g, 0.735mol) was added slowly (lhr). The temperature was kept at 10-15 0 C during 10 the addition. The reaction mixture was left overnight at room temperature. TLC [silica, chloroform/methanol/anmmnonia(33%) 90:10:1 (v/v)] showed one spot at Rf -0.4. The product, N-Pyridin-4-ylmethyl - acetamide was not isolated and was processed to the next step. Part of the reaction mixture (43ml) was evaporated. The acetamide salt was obtained as yellow oil (14.5g). Part of the oil (1.9g, 50.06mol) was dissolved in methanol (40ml), stirred under nitrogen 15 atmosphere and protecred from light. Iodomethane-d 3 (10g, 0.07mol) was added, maintaining the temperature of the reaction mixture at 15-25 0 C. The reaction mixture was left overnight at room temperature then triturated twice with ether (2x200ml). 4-(Acetamido- methyl)- 1-methyl d 3 -pyridiniumm iodide was obtained as yellow solid TLC [silica, chloroform/methanol/ammonia(33%) 90:10:1 (v/v)] showed one spot at Rf -0.05] and was reduced in the next step without further 20 purification. To a cold (ice-water bath) solution of the pyridinum iodode salt in methanol (40ml) under nitrogem atmosphere, sodium borohydride (3.9gr, 0.2mol) was gradually added (2hir) so the temperature was maintained at 15-30 'C, the reaction mixture was stirred for additional 2hrs at room temperature and left overnight without stirring. The solvent was evaporated. N-(I-methyl 25 d 3 -1,2,3,6,-tetrahydropyridine- 4 -ylmethyl)- acetamide was obtained as stick yellowish oil (8.5 g). 1 H-NMR (CDC1 3 ) 5 1.98 (s, 3H, CH 3 CN), 2.12 (min, 2H11), 2.52 (t, 2H11), 2.91 (min, 2H), 3.76 (d,
CH
2 NHCO), 5.52 (min, CH=C), 6.66 (br. s, NH) ppm. MS m/e 172(M). In a three-necked round bottom flask (250ml) equipped with an addition funnel and condenser with a calcium chloride tube on its top, a solution of the tetrahydropyridine acetamide (8.5gr) in dry 30 acetonitrile (70ml) was added. To the stirred solution, phosphorous pentasulfide (6.7gr, 0.03mol) was added followed by triethylamine (12gr, 0.12mol) which was added from the additional funnel during 10-15min. The obtained solution was heated under reflux for 5hirs and then left at 15 oC for three days. The solution was evaporated, basified with 10% aqueous potassium carbonate, then extracted with WO 03/092580 PCT/IL03/00357 46 chloroform. The organic phase was dried and evaporated. Crude black N-(1-methyl d 3 -1,2,3,6,-tetrahydropyridine- 4-ylmethyl)- thioacetamide (6.62gr, 0.036mol) was obtained. 'H-NMR (CDC13) 6 2.21(m, 2H), 2.56(s, CH 3 CS), 2.58(t, j = 4.4Hz, 2H), 2.97(m, 2H), 4.23(d, j = 4.4Hz,
CH
2 NI-I), 5.65(m, CH=C), 7.41(br s, NH) ppm. MS m/e 187(M). 5 In a flask (150ml), polyphosphoric acid (30g) was added to the crude thioacetamide (6.5gr). The reaction was stirred and heated to 170 0 C for 3.5h. The hot reaction mixture was slowly poured into a stirred 25% aqueous sodium carbonate (150ml). 25% Aqueous sodium carbonate (50ml) wasadded to the residue in the reaction flask and the two solutions were combined. The basicity was raised by addition of an 50% aqueous sodium hydroxide (6ml) and the reaction mixture was extracted with 10 chloroform. The organic phase was separated, dried and evaporated. The residue was dissolved in petroleum ether (100ml) to give after 12h at -20 0 C a solution and a small amount of insoluble material. The petroleum ether solution was evaporated and the obtained oil (Sg) was distilled at reduced pressure (b.p. 50-53'C, 0.2mmHg) to give AF402 (2.15g, 0.012mol). 1 H-NMR (CDCl 3 ) S 1.87(m, 2H), 1.94(m, 2H), 2.1(m, 2H), 2.2(t,j = 1.7Hz, 3H), 2.76(m, 2H), 3.92(m, 2H) ppm. MS m/e 187(1M). 15 EXAMPLE 18: Synthesis ofN-[(2.8-Dimethyl-1-oxa-8-aza-spirof4.51dec-3-vlidene)-methyl-aminel-N-oxide, AF600 0
O
0 N AF600 To a solution of N-methylhydroxylamine hydrochloride (0.85gr, 0.01mol) in ethanol (13.5ml) 20 was added sodium acetate (0.84gr, 0.01mol). A white precipitate was obtained, a solution of 2,8-dimethyl-1-oxa-8-aza-spiro[4.5]decan-3-one (1.62gr, 0.0088mol) in ethanol (7ml) was added and the mixture was stirred at room temperature for 4.5hrs. The solvent was evaporated, dichloromethane (675ml) was added and the obtained solution was washed with 20% aqueous sodium carbonate. The organic phase was dried, the solvent was evaporated and flash chromatography (silica, 25 CHC1 3 /MeOH/NH 4 0OH 90/10/1) gave AF600 (1.5gr) as a mixture of two isomers [less polar isomer(A)/more polar isomer(B) 1:4]. 1 H-NMR (CDC1 3 ) 5 1.45 [d, j = 6.37Hz, CH 3 (A)], 1.54 [d,j = 6.48Hz, CH 3 (B)], 1.7-1.9 (m), 2.29 [S, NCH 3 (A)], 2.31 [s, NCH 3 (B)], 2.32-2.5 (m, 3H), 2.62 (s, CH 2 ), 3.63 [s, CH 3 NO (A)], 3.68 [s, CH 3 NO (B)], 4.78 [br CH (A)], 4.85 [br CH (B)] ppm. MS m/e 212 (M), 196, 169, 126, 110, 96,70. 30 WO 03/092580 PCT/IL03/00357 47 EXAMPLE 19: Synthesis of N-(2. 8-Dimethyl-l-oxa-8-aza-spiro4.51dec-3-ylidene)-benzyl-aminel-N-oxide. AF604 O0 Ph 0 N AF604 To a solution of N-benzylhydroxylamine hydrochloride (1.27gr, 0.008mol) in ethanol (6ml) was 5 added sodium acetate (0.65gr, 0.008mol). A white precipitate was obtained. A solution of 2,8-dimethyl-1-oxa-8-aza-spiro[4.5]decan-3-one (1.3gr, 0.0072mol) in ethanol (3.5ml) was added and the mixture was stirred at room temperature for 4hrs. The solvent was evaporated, dichloromethane (450ml) was added and the obtained solution was washed with 20% aqueous sodium carbonate. The organic phase was dried, the solvent was evaporated and flash chromatography (silica, CHCl 3 / 10 MeOH/NIH4OH 90/10/1) gave AF604 (1.81gr) as a mixture of two isomers [less polar isomer(A)/more polar isomer(B) 1:8]. 'H-NMR (CDC1 3 ) 8 1.42 [d,j = 6.35Hz, CH, (A)], 1.53 [d,j = 6.45Hz, CH 3 (B)1, 1.7-1.9 (m), 2.29 (S, NCH 3 ), 2.32-2.55 (m), 2.62 (m, CH 2 ), 4.85(br CH), 4.91[s, CH 3 NO (A)], 4.97 [s,
CH
3 NO (B)] ppm. MS m/e 288 (M), 272, 254, 197, 153, 91(100%). 15 EXAMPLE 20: Synthesis of N-[(2.8-Dimethyl-_ 1-oxa- 8-aza- spiroR4.51dec- 3-lidene)-isopropvl-amine]- N-oxide, AF605 0 N
O
0o 4 N AF605 To a solution of n-isopropylhydroxylamine hydrochloride (1.84gr, 0.01mol) in ethanol (7ml) was 20 added sodium acetate (0.91gr, 0.011mol). A white precipitate was obtained. A solution of 2,8-dimethyl-1-oxa-8-aza-spiro[4.5]decan-3-one (1.84gr, 0.01mol) in ethanol (4ml) was added and the mixture was stirred at room temperature for 4.5hrs. The solvent was evaporated, dichloromethane (500ml) was added and the obtained solution was washed with 20% aqueous sodium carbonate. The organic phase was dried, the solvent was evaporated and flash chromatography (silica, 25 CH 2 C1 2 /MeOH/NH 4 OH 90/10/1) gave AF605 (1.5gr) as a mixture of two isomers [less polar WO 03/092580 PCT/IL03/00357 48 isomer(A)/more polar isomer(B) 1:4]. 1H-NMR (CDC1 3 ) 5 1.40 (d, j = 6.6Hz, CH 3
CH
2 ), 1.42 [d, j = 6.2Hz, CH 3 (A)], 1.6-1.8 (m), 2.29 [s, CH 3 (A)], 2.31 [s, CH 3 (B)], 2.35-2.57 (m, 3H), 2.63-2.71 (mn, 2H,
CH
2 ), 4.05 [m, CHNO (A)], 4.18 [m, CHNO (B)], 4.83 (m, 1H, OCH) ppm. 5 EXAMPLE 21: Synthesis of N-f(2-Ethyl- 8-methyl- 1-oxa- 8-aza- spirof4.51dec- 3-vlidene)-methyl-amninel- N-oxide, AF601 O0 0 N AF601 To a solution of N-methylhydroxylamine hydrochloride (0.95gr, 0.011mol) in ethanol (14.3ml) 10 was added sodiumacetate (0.94gr, 0.011mol). A white precipitate was obtained, a solution of 2-ethyl-8-methyl-1 -oxa-8-aza-spiro[4.5]decan-3-one (2.1gr, 0.01mol) in ethanol (7.4ml) was added and the mixture was stirred at room temperature for 5.5hrs. The solvent was evaporated, dichloromethane (770ml) was added and the obtained solution was washed with 20% aqueous sodium carbonate. The organic phase was dried, the solvent was evaporated and flash chromatography (silica, 15 CHCl 3 /MeOH/NH 4 OH 90/10/1) gave AF601 (2.4gr) as a mixture of two isomers [less polar isomer(A) and more polar isomer(B)]. 1 H-NMR (CDC1 3 ) 0.94 [t, j = 7.4Hz, CH 3
CH
2 (B3)], 0.99 [t, j = 7.4Hz,
CH
3
CH
2 (A)], 1.59 (m, 1H1), 1.74-1.86 (m), 1.98-2.05 (m, 2H), 2.29 [s, NCH 3 (A)], 2.30 [s, NCH 3 (B)], 2.45-2.60 (m, 5H), 3.63 [s, CH 2 (A)], 3.69 [s, CH2 (B)], 4.66 [br OCH (A)], 4.78 [br OCH (B)] ppm. MS m/e 226 (1), 209, 197, 181, 169, 152, 138, 126, 110, 96,70. 20 EXAMPLE 22: Synthesis of N-[(2-methyl-8-phenvl-1-oxa-8-aza-spirof4.5]dec-3-v1idene)-methyl-amine]-N-oxide. AF602 O0 Ph N_ 0 N AF602 AF602 WO 03/092580 PCT/IL03/00357 49 To a solution of N-methylhydroxylamine hydrochloride (0.33gr, 0.004mol) in ethanol (5.4ml) was added sodiumacetate (0.32gr, 0.004mol). A white precipitate was obtained, a solution of 8-methyl-2-phenyl-1 -thia-4,8-diaza-spiro[4.5]decan-3-one (0.85gr, 0.004mol) in ethanol (3ml) was added and the mixture was stirred at room temperature for 4.5hrs. The solvent was evaporated, 5 dichloromethane (250ml) was added and the obtained solution was washed with 20% aqueous sodium carbonate. The organic phase was dried, the solvent was evaporated and flash chromatography (silica, CHC1 3 /MeOH/NH 4 0H 90/10/1) gave AF602 (130mg) as a mixture of two isomers [less polar isomer(A)/more polar isomer(B) 1:3]. 1 H-NMR (CDC13) 6 1.24 (m, CH 2 ), 1.7-1.97 (m), 2.29 [s, NCH3 (A)], 2.31 [s, NCH 3 (B)], 2.32-2.5 (m), 2.72 (m, CH 2 ), 3.30(m), 3.67 [s, CH 3 NO (B)], 3.71 [s, CH 3 NO [0 (A)], 5.49 [br CH (A)], 4.74[br CH (B)], 7.28-7.56(m, ArH) ppm. MS m/e 274 (M), 257(100%), 245, 168, 112,96, 70. EXAMPLE 23 Synthesis of Dihydro- 5'-methylspiro[l-azabicyclof2.2.2]octane- 3,5'-(4'H)-3'-vlidene- methylamine 15 N-oxide, AF603 0o NN +-0 N AF603 To a solution of N-methylhydroxylamine hydrochloride (1.17gr, 0.014mol) in ethanol (15ml) was added sodium acetate (1.15gr, 0.014mol). A white precipitate was obtained. a solution of dihydro-5'-methylspiro[1-azabicyclo[2.2.2]octane-3,5'-(4'H)-3'-one (2.1gr, 0.01mol) in ethanol 20 (7.4ml) was added and the mixture was stirred at room temperature for 4.Shrs. The solvent was evaporated, dichloromethane (770ml) was added and the obtained solution was washed with 20% aqueous sodium carbonate. The organic phase was dried, the solvent was evaporated and flash chromatography (silica, CHC13JMeOH/NH4OH 90/10/1) gave AF601 (0.45gr) as a mixture of two isomers [less polar isomer(A) and more polar isomer(B)]. 1H-NMR (D 2 0) 5 1.22 [t, j = 7.09Hz, CH 25 (B)], 1.46 [t,j = 7.0Hz, CH (A)], 1.35 [d,j = 6.5Hz, (C1H3) 2 CI-], 1.41-1.58 (m), 1.57-1.9 (m), 2.59 (m), 2.67-3.01 (m), 3.01-3.26 (m), 3.47 [s, N(O)CH 3 (A)], 3.49 (m), 3.52 [s, N(O)CH 3 (B)] ppm. MS m/e 224 (M), 207,195, 178, 138, 124, 96, 83 (100%).
WO 03/092580 PCT/IL03/00357 50 EXAMPLE 24: Synthesis of 2-Ethyl-8-methyl-l1-thia-4.8-diaza-spirof4.5]decane-3-thione. AF510 S S NH N I AF510 A mixture of AF267 (214mg, Immol) and Lawesson's Reagent (280mg, 0.692mmol) in 5 acetonitrile (5 ml) was heated under reflux for 17 hrs. The solvent was removed and residue was dissolved in concentrate aqueous sodium carbonate (0.5ml) and then extracted with ethyl acetate. The extract was dried and the solvent evaporated. The residue (250mg), recrystallized first from toluene and then from acetonitrile gave pure AF510. 'H-NvMR (CDCl 3 ) 5 1.03 (t, CH 3 CH2), 1.83 (mn), 1.93-2.44 (min), 2.30 (s, CH 3 N), 2.80 (m, 2H1), 4.21 (dd, SCH), 8.66 (br, NH) ppm. MS m/e 230 (V), 197 0 (1+-SH), 156, 128, 96 (100%). EXAMPLE 25: Synthesis of 4-Benzvl-8-methyl-1 -thia-4.8-diaza-spiro[4.51decan-3-one, AF282 0 S N N I AF282 15 In a three-necked flask equipped with a magnetic stirrer and Dean-Stark and two dropping funnels a solution of mercaptoacetic acid (8ml, 0.242mol) in benzene (75ml) was heated to reflux. Benzyl bromide (13ml, 0.242mol) and 1-methyl-4-piperidone (9.3ml, 0.08mol) were added simultaneously dropwise (45min) and the reaction mixture was refluxed for additional 1.5h (2ml of water were collected). The reaction mixture was cooled to room temperature, water (30ml) was added 20 and the organic phase was separated, dried and the solvent was evaporated. Flash chromatography (silica, 10% methanol in chloroform) of the residue gave AF282 (3.8g). 'H-NMR (CDC13) 51.65 (inm, 2H), 2.19 (m, 4H), 2.27 (s, 3H, NCH 3 ), 2.78 (m, 2H), 3.64 (s, 2H, SCH 2 ), 4.78 (s, 2H, NCH 2 ), 7.25 (SH, Ar) ppm.
WO 03/092580 PCT/IL03/00357 51 EXAMPLE 26: Synthesis of 4 -(2.4-Dimethoxvbenzvl)-8-methyl-1 -thia-4. 8-diaza-spiror4.51decan-3-one. AF286 0 S N OMe N MeO I AF286 In a three-necked flask equipped with a magnetic stirrer and Dean-Stark a solution of 1 5 methyl-4-piperidone (0.27ml, 2.4mmol), 2,4-dimethoxybenzylamine hydrochloride (0.72gr, 3.Smmol)and mercaptoacetic acid (0.24ml, 3.5mmol) in benzene (Sml) was refluxed for 3h. The reaction mixture was cooled to room temperature, water (10ml) was added and the organic phase was separated. The pH of the aqueous phase was adjusted to pH 10 with 2.5N aqueous sodium hydroxide solution and then the aqueous phase was extracted with chloroform. The organic phases were 10 combined, dried and the solvent was evaporated. Flash chromatography (silica, 10% methanol in chloroform) of the residue gave AF286 (140mg, 15%yield).'H-NMR (CDCl 3 ) 81.66 (min, 2H), 2.21 (m, 4H), 2.27 (s, 3H, NCH 3 ), 2.77 (min, 2H), 3.64 (s, 2H, SCH 2 ), 3.78 (s, 3H, OCH 3 ), 3.80 (s, 3H, OCH 3 ), 4.52 (s, 2H, NCH 2 ), 15 EXAMPLE 27: Synthesis of 4-(tert-Butvloxvcarbonyl)-8-methyl- 1 -thia-4.8-diaza-spirof4.51decan-3-one, AF284 0 S N-BOC N AF284 To a solution of AF277 (2.60gr, 13.97mmol) in dichloromethane (60ml), triethylamine (1.95m1, 13.99mmol) di-tert-butyl dicarbonate (3.85ml, 16.76mmol) and 4-dimethylaminopyridine (1.71gr, 20 13.99mmol) were added. The obtained solution was stirred overnight at room temperature then the solvent was evaporated. Flash chromatography (silica, CHC1 3 /MeOHNIN 4 OH 80/20/1) of the residue gave AF284 (4gr, >95% yield). 'H-NMR (CDC1 3 ) 6 1.55 (s, 9H, OC(CH1 3
)
3 ), 1.70-1.82 (min, 2H), 2.26 (mn, 211), 2.28 (s, 3H, NCH 3 ), 2.83-2.87 (mi, 4H), 3.53 (s, 2H, SCH 2 ) ppm. 25 EXAMPLE 28: Formulation ofAF 150O(S) in paraffin oil + stability studies The stability of 10% w/w AF150(S) in pharmaceutical acceptable paraffin oil (Paraffin oil Eur. Ph) was examined at 40 0 C under air or nitrogen atmosphere, in the presence or absence tocopherol.
WO 03/092580 PCT/IL03/00357 52 Samples were analyzed after two months storage. TLC, FHPLC and GC were used to detect and determine the quantity of possible degradation product(s). Color changes were determined in comparison with Paraffin oil with or without tocopherol under the same conditions. AF150(S) was stable in the paraffin oil formulation. Degradation products, the thiolamide derivative (M 202) 5 obtained by hydrolysis of AF150(S), above 0.1% were not detected in any tested samples. A slight yellow color was observed in samples without tocopherol but color was not developed in samples containing 0.5% w/w tocopherol in AF150(S). EXAMPLE 29: .0 AF 150O(S) citrate salt + stability studies To a solution of AF150(S) (30.13gr, 163.8 mmol) in 2-propanol (60 ml) and tetrahydrofuran (100 ml) was added dropwise over 45 min a solution of anhydrous citric acid (29.51gr, 153.5 mmol) in 2-propanol (200 ml). The resulted mixture was stirred at room temperature under argon atmosphere for additional 2h, then the resulted white precipitate was filtered and washed with hexane under argon L5 atmosphere. The white solid was introduced into a drying pistol which contained P 2 0 5 , the drying pistol was evacuated (0.2 mmHg) temperature and then heated at 55-600C for 6h. AF150(S) citrate (53.8 g, 87.6% yield) was obtained. TLC (2% NH4OH in methanol) Rf 0.57; mp. 146.5-147.5 oC; 1H NMR (300MHz, D 2 0-Na 2
CO
3 , pH 12) 8 1.80(m, 4H), 2.08 (s, 3H, CH 3 C=N), 2.17(s and m, 5H,
CH
2
+CH
3 N+), 2.46 (ABq, j=15.2Hz, 4H, 2CH 2
CO
2 H), 2.73(m,2H), 3.81(s, 2H, CH 2 N=C) ppm. 3C 20 NMR (300MHz, D 2 0-Na 2
CO
3 , pH 12) 19.77, 36.17, 44.16, 45.64, 53.08, 72.71, 75.03, 170.91, 179.19 and 181.86 Hz. HPLC analyses of samples of this salt stored under various conditions (at 60 0 C for three months; in air at room temperature for three months) compared to a reference standard stored under anhydrous conditions showed the salt to be highly stable." 25 EXAMPLE 30: Brain penetration of compounds 1. pKa of AF267B: The free-base (non-ionized) form of the compound AF267B crosses the brain blood 30 barrier. Since AF267B has a pKa of 7.8, at the pH = 7.35 of the cerebrospinal fluid (CSF), 26.2% of the compound are in a free base form, calculated as shown below. This indicates that AF267B is highly penetrable into the brain since the free base is the specie that crosses the blood brain barrier. In comparison, some other known pharmaceutical CNS active compounds, for example wherein the base is quinuclidinyl, have a pKa > 9 (where the tertiary amine is highly basic). For such compounds at the 35 relevant pH of 7.35, only 2.2 % are in non-ionized form. This indicates a higher preference for the brain for AF267B vs. such compounds. These calculations are based on the following: WO 03/092580 PCT/IL03/00357 53 BH - B + I-F % B=100-% BI- = 1 0 0 -1 0 0
/[
1
+
1 0 (pH-P K a )] pKa (AF267B) =7.8 pH (CSF) =7.35 5 % B=100-100/1+10- 4 5 ] % B =26.2% For a base B' having a pKa = 9.0 % B'=100-100/1+10- 165 ] % B'=2.2% 10 2) Rats were treated with AF267B (2 mg/kg, po) and plasma vs. brain levels of the drug were analyzed by GC. It was found that AF267B has a preference for the brain vs plasma: a) by comparing the area under the curve extrapolated to infinite time (AUC) both in whole brain (ng/gr)*hr vs plasma (ng/ml)*hr both iv (1mg/kg) and po (2 mg/kg), respectively, it was found 15 that the ratio of AUC brain/AUC plasma is: for males 1.79 (iv) and 2.43 (po) and for females 1.32 (iv) and 1.25 (po). Thus greater amounts of the compound are found in the brain than in plasma; b) by comparing the ratio of Cmax brain (ng/g)/Cmx plasma (ng/ml) (po) it was found that 25% of the compound in males and 16% in females is found in the brain. This calculation was based on the 20 brain weight (2gr) vs. total plasma volume (14 ml). This also indicates a high percentage of the compound in the brain. 3) ex vivo studies of AF150(S) and AF267B (100 ptmole/kg, po) in mice brain tissue (GC analysis of AF150(S) or AF267B vs a standard compound (AF261) added to the brain tissue, or by displacement of 25 a radioactive muscarinic compound such as tritiated-oxotremorine-M from the brain tissue) also show clearly a high brain penetration vs. plasma (GC and binding studies). AF150(S): Tmax=1-10min; Tin 2 =21 and 53 min (two phases), iv; MRT (mean retention time) = 50 min, iv, po. AF150(S) has a fast brain penetration (1 min iv); Cmax = 40.7 gmole/kg (40.7 % in brain from the amount administered po); AF267B: detected in the brain between 2-240 min after dosing, a peak at 20-30 min, MRT=128 min; 30 Cmax= 36.4 [mole/kg (36.4 % in brain from the amount administered po). EXAMPLE 31 Detection of AF292 following AF267B administration to Beagle dogs. The purpose of this study was to determine the levels of AF267B and AF292 (a metabolite of 35 AF267B) in dog plasma following 13 weeks subchronic singly daily administration of AF267B (1.5, 3 and 6 mg/kg, po to male and female dogs) according to the method (see below).
WO 03/092580 PCT/IL03/00357 54 Internal Standard (AF261): 2-Methyl-8-methyl- 1-thia-4,8-diaza-spiro[4.5] decan-3-one; MW 200. The analytical plasma samples originate from the in-life part of this study. Concentrations of AF267B and AF292 were determined by: Column: Purospher STAR RP18e (4 x 50 mm, 3 gm) 5 Mobile Phases: Solvent A: 1 g/1 (HN 4
)
2
CO
3 (H20) Solvent B: Methanol Loop/Injection Volume: 50 .pl/10 pl lonisation Mode: Atmospheric Pressure Chemical Ionisation (APCI); positive ions Sheath Gas Pressure: Nitrogen: 70 psi 10 Capillary Temperature: 250 oC Spray Voltage: ~ 3.6 kV Detection Mode: SRM (selected reaction monitoring) AF292: m/z: 201.0 [CE (CE = collision energy 30V)] -> m/z: 70.0 (0.0-5.2 min)] AF261: m/z: 201.0 (CE 35V) -) m/z: 70.0 (5.2-6.2 min); Internal standard 15 AF267B:m/z: 215.0 (CE 30V)-4 m/z: 70.0 (6.2-10.0 min) Collision Gas (CID): 2.5 mTorr/Argon Results: After 13 weeks of repeated daily dosing AF267B has a plasma half life of 1-2 hrs with Tmax 20 =1.5-3 h, Cmax (ng/ml)= 162-1352 (linearly dose-dependent) and AUC(o-0t) (ng*h/ml) = 712-3947 (linearly dose-dependent). AF292 has an approximately ten fold longer plasma life (~9-20 hrs) with Tm ax = 3 hrs, Cmax (ng/ml)=136-555 and AUC(o) (ng*h/ml) = 616-2451 (linearly dose-dependent). In comparison to AF267B, the pharmacokinetic profile of AF292 can be summarized as follows: AF292 has a T 1 /2 in plasma about 3-5 times longer than AF267B (for example, AF292 T 1 2 =10.6 hr for 25 females, versus T 1 /2 = AF267B in females). The Cm,,, of AF292 is 50-90% vs. the Cmax of AF267B. AF292 shows an apparent shift to the right of Cmax vs. the Cm of AF267B (due to a delay in the appearance in plasma of AF292 vs. AF267B). On the basis of this observation, it will be appreciated that AF267B and AF292 may together form a pharmaceutical combination with a longer plasma T 1 /2 than either compound alone. Such a combination may be adminstered as such. 30 EXAMPLE 32 Effects of the tested compounds on secretion of a-APP, in cell cultures stabily transfected with the M1 mnAChR and in rat primary hippocampal and cortical neuronal cultures. Cells were plated in 6 well culture plates and used at the age of 3-5 days after plating. Cells were 35 washed twice in serum-free medium and incubated for 1 hour at 37 0 C with AF150(S) and AF267B or AF292. The cell cultures were exposed for 1 hr to various concentrations of these tested compounds (10
-
6 -10 - 3 M), and to carbachol (10 -4 M). Cells exposed to medium alone are referred as controls. Carbachol, rivastigmine and deprenyl were used as reference compounds.
WO 03/092580 PCT/IL03/00357 55 Cell supernatants were collected into Eppendorff tubes containing a cocktail of protease inhibitors (5 units/ml aprotinin, 5 mg/ml pepstatin A, 5 mg/ml leupeptin and 104 M Phenylmethylsulfonylfluoride (PMSF, a protease inhibitor); Sigma, USA). The collected media were concentrated with Centricon tubes (Amicon, Beverly, MA, USA) and kept frozen for a-APPs secretion 5 determination. Equal amounts of protein (50-100jLg) were loaded and separated on 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Followed by western blotting onto nitrocellulose membrane, blocked by fat-free milk, and probed for 24 hours at 40C, with anti-Alzheimer's precursor protein, A4 monoclonal antibody 22C11 (0.25 p.g/ml; Boehringer Mannheim, Germany). The nitrocellulose membranes were washed and incubated for 2 hours at room 10 temperature with peroxidase-linked goat anti-mouse IgG antibodies (Jackson Immunoresearch, USA), followed by extensive washout and staining with enhanced chemiluminescence detection system (Amersham). Quantitative evaluation of the immunoreactive bands, on the exposed films, was performed by video-imaging densitometry (Gel-aid software; Galai Co., Israel). APPs levels were expressed as x-fold increase over basal levels. In cell cultures stably transfected with M1 mAChR, 15 a-APP, -induced secretion was also calculated as % of maximal response to 104M carbachol. ca-APP, secretion in cells expressing the M1 mAChR increased dose-dependently following agonist stimulation. The maximal increase was obtained at 10 - 4 0 M AF267B (about 6-fold increase over control) and was equal to the maximal increase elicited by carbachol. Addition of the muscarinic antagonist, atropine at 10 5 M, inhibited completely the secretion of APP, induced by AF267B, 20 indicating that the effect of AF267B is mediated via M1 mAChR. In several experiments, the activity of AF267B was compared to.the activity of AF150(S). The results show that AF267B is more efficacious and potent than AF150(S) (50% of maximal response of carbachol for AF150(S) vs equal to maximal response of carbachol for AF267B). Additionally AF267B is more efficacious and more potent than its racemate (AF267) or AF102B (50% of maximal response of carbachol) on a-APPs secretion, while the 25 less potent enantiomer AF267A is similar in potency to AF102B. AF292 was as effective as AF267B (EC50=3p.M) and carbachol in activating elevation of APP,, while rivastigmine and deprenyl were not effective in elevating APP, levels. Addition of atropine (10 gM) inhibited the secretion of APPs induced by carbachol, AF267B and AF292, indicating that the effect of these agonists is mediated via M, mAChR. 30 Taken together, these results show that AF267B is a selective Ml muscarinic agonist and a "drug-prodrug" for AF292, which itself is a selective Ml muscarinic agonist and a weak M3 muscarinic antagonist. AF267B and AF292 together form a pharmaceutical combination with a longer plasma half life and longer muscarinic activity than either compound alone. Using the above tests, AF700 and AF704 were also found to be effective in increasing APP, 35 levels in this preparation (at 100 AM 50% of maximal effect of carbachol). The effect of various muscarinic agonists on the levels of secreted APP, were followed using rat primary cell cultures prepared from hippocampus, cerebral cortex (both which contain mainly Ml mAChR) and spinal cord (which contains M2 receptors). In this study the effects of carbachol (a non-selective muscarinic agonist), oxotremorine (> M2 selective muscarinic agonist), physostigmine (a WO 03/092580 PCT/IL03/00357 56 cholinesterase inhibitor) and AF102B, AF150(S) and AF267B (Ml selective muscarinic agonists) on APPs secretion were tested.] Primary cell cultures were prepared from embryos of Sprague-Dawley rats. The experiments were performed with cultures of hippocampus, cerebral cortex and spinal cord following the guidelines 5 "Guide for Care and Use of Laboratory animals", National Research Council, Washington, DC 1996. Brain tissues, hippocampus, cerebral cortex and spinal cord were removed from 13-14 or 18-19-day-old rat fetuses, respectively, by free-hand dissection and transferred into cold Gey's Balanced salt Solution (Gibco, BRL) containing 6 mg/ml glucose. After removal of meninges, the dissected tissue was mechanically dissociated using Pasteur pipettes followed by tripsyn-DNAase solution to obtain cell 10 suspension. Dissociated cells were transferred to Dulbeco's Modified Eagle Medium (Biological Ind. Beit-Haemek, Israel) containing: 6 mg/ml glucose; 2 mM L-glutamate, 1000 IU/ml penicillin. The cell suspension was plated on poly-L-lysine (1 mg/ml)-pre-coated 12-well culture tissue plates at a density of 4X10 5 and 6X105 cells/well for hippocampal and cortical cells, respectively. Cell cultures were maintained for about 2 weeks in 37'C incubator (95% air & 5% C0 2 ). Cells at 11-14 days in vitro were 15 extensively washed and then subjected to various treatments as detailed below. Hippocampal and cortical cells were incubated with the tested ligands at a concentration of 100 ptM for 1 h in magnesium-free Locke-HIEPES buffer consisting of: 154 mM NaC1, 5.6 mM KC1, 3.6 mM NaHCO 3 , 1.3 mM CaC1 2 , 5.6 mM glucose and 10 mM HEPES, pH 7.4, containing 0.02% BSA. In the blockade studies the muscarinic agonists were co-incubated with the antagonist, pirenzepine (10 riM). Cells 20 exposed to buffer alone were referred to as control. At the end of the incubation period, the conditioned media was removed and transferred to Eppendorff tubes, which contained a cocktail of protease inhibitors (as specified above). The supernatants were concentrated by centrifugation (2,500Xg for 45 min at 4oC) using Centricon-30 concentrators (Amicon, Inc. MA USA) and frozen at -70 0 C till APPs levels were determined. 25 The content of protein in samples was determined in microplates according to Bio-Rad assay. Equal protein amounts of each sample (, 40 Rg/lane) were loaded on 10% SDS-PAGE. When electrophoresis was completed, gels were blotted onto nitrocellulose membranes, blocked by fat-free milk and APP, bands were probed using the anti-Alzheimer precursor protein A4 (monoclonal 22C11, Boehringer Mannheim) and the secondary probe peroxidase-linked rabbit anti mouse IgG (Jackson 30 ImmunoResearch, P). Following extensive washout the bands were stained with TMB (single solution, Zymed Lab., California) or developed with the Renaissance Chemiluminescence Reagent (DuPont, NEN) followed by exposure to an autoradiography film (Hyperfilm-ECL, Amersham). Quantitative determination of the total APPs bands was performed by video-imaging densitometry (Gel-aid software, Galai Co. Israel). Data obtained for APP, were expressed as fold increase over control where the control 35 was cells incubated with Locke buffer alone. Primary rat cortical and hippocampal cultures cell cultures were exposed to the non-selective agonist carbachol (CCh) and oxotremorine (>M2 selective), to the M1 muscarinic agonists, AF150(S) and AF267B and to the cholinesterase inhibitor, physostigmine, all at 100 ptM. The Ml agonists induced a significant increase in APPs secretion in both cell systems used, hippocampus and cortex as WO 03/092580 PCT/IL03/00357 57 compared to levels determined in control cell cultures. In cortical cell cultures the increase in APPs levels ranged from 2.5 to 3.1-fold increase over control and an increase in the range of 1.8-2.8-fold over control was found in hippocampal cell cultures. AF150(S) and AF267B were more potent than CCh (2.8-fold and 1.5-fold over control, respectively). Oxotremorine and physostigmine were inactive. 5 APP,-induced secretion by AFl50(S), AF267B and CCh was completely blocked by the Ml selective antagonist, pirenzepine (10 rLM). These agonists did not activate APPs secretion in the spinal cord cultures, as these neurons do not contain M1 mAChR. EXAMPLE 33 .0 Neurite-outgrowth response to muscarinic agonists in the absence or presence of neurotrophins Rat pheochromocytoma cells transfected with M1 mAChR cells were grown as described in Gurwitz et al, (NeuroReport 6, 485,1995). For determination of neurite outgrowth, cells plated in six-well plates were used 3-5 days after plating. Growth factors were added 1 day after plating and muscarinic agonists were added for the last 24 hrs. 15 Cells were observed under an inverted microscope. The percent of cells with neurite longer than cell diameter were scored in three random fields of several hundred cells from each well. Results were expressed as a percent of cells with neurites. Treatments were performed in triplicate cells. Both NGF (50ng/ml) and EGF (100ngiml) were added 1 day after plating. Muscarinic agonists were added 24 hours before scoring. 20 The neurotrophic-like effects of AF102B, AF150(S) and AF267B vs carbachol (CCh) and their interaction with neurotrophins such as NGF, basic fibroblast growth factor (bFGF) and epidermnal growth factor (EGF) were evaluated. Maximal response to CCh was 80% compared to 60% for AF267B and 30% for AF150(S). Pretreatment of rat pheochromocytoma cells transfected with Ml mAChR cells with NGF synergistically augments the neurotrophic response to all ligands tested and the 25 efficacy of the agonists tested was increased. There is an observable difference between the cellular response of rat pheochromocytoma cells transfected with M1 mAChR cells to NGF and bFGF on the one hand (induce differentiation) and to epidermal growth factor, EGF on the other hand (induces proliferation). The proliferating profile of EGF changed in the presence of muscarinic agonists as EGF together with muscarinic agonists 30 induced an accelerated differentiation. Taken together, the above results show that M1 selective agonists, alone or in combination with either endogenous or exogenously administered growth factors, may be used to induce neurotrophic effects beneficial in the treatment of neurodegenerative disorders, such as AD. AF292, AF700 and AF704 were also found to be neurotrophic, and this effect was blocked totally 35 by atropine, indicating the mucarinic nature of the effect.
WO 03/092580 PCT/IL03/00357 58 EXAMPLE 34 Effects on A3 levels in vitro Primary mixed rat cortical neurons infected with recombinant Semliki Forest virus encoding either the human APP695 or APP C99 or Cl1 (Fassbeder et al, PNAS,98: 5856, 2001), were treated 5 with one of the test compounds (AF102B, AF150 or AF267B, respectively for 5-8 hours. In these cell cultures APP is cleaved by y- and P3-secretase to produce AP3, whereas cX-secretase destroys Ap3. However, ac-secretase is not present intracellularly. Cells were lysed and A3 precipitated with W02 (anti AP3 antibody). The mechanism of Af3 modulation was tested using C99 (and C 111) that are truncated constructs 10 generated from APP. Unlike APP, C99/C 111 are direct substrates for y-secretase. With both constructs it is possible to directly assay for y-secretase activity, while with APP this is not possible. Both constructs are - as compared to APP - inefficient substrates for ca-secretase. Synergistc effects were also evaluated with CDX (methyl-p3-cyclodextrin), an agent which extracts cholesterol from the plasma membrane. CDX inhibits also AP3 production. CDX-treated cells were treated in addition to the 15 respective muscarinic agonist for 5 min to reduce the cholesterol content. AP3 levels were reduced upon treatment with the muscarinic agonists both in the cell lysate and medium in this system. AF267B was at least 5-fold more potent than AF150(S) in decreasing AP3 levels, being active in the tM range. A synergistic effect between AF267B and the general cholesterol lowering agent, CDX (5mM), in their efficacy to decrease AP3 in this system to undetectable A3 levels, 20 was observed. It was also observed in these studies that the present M1 agonists, in addition to activating ac-secretase, inhibit y-secretase. AF267B reduced the release of Af3-like fragments (all fragments being in the 3-4 Kda) range by approximately 50%. This is equivalent to a y secretase activity reduction of 50%. This was also evidenced by a complete loss of the p3 fragment (a fragment of APP resulting from y-secretase cleavage) in the AF267B (lmM)-treated cells vs the control. No other 25 compounds have been reported with such a combined beneficial property on the various secretases (x-, P3-, and y--). The results indicate that the combination of an Ml agonist with a cholesterol lowering agent, such as a statin, enables the lowering of the dosage of the Ml agonist and thus reduction of possible side effects of the Ml agonist. 30 EXAMPLE 35 AF267B decreases elevated 3-amvloids in cortex in hvpercholesterolemic rabbits Dietary cholesterol induces Alzheimer-like Ap-immunoreactivity in rabbit brain (Sparks et al. Exp Neurol 1994; 126:88-94; Sparks Nutr Metab Cardiovasc Dis 1997; 7:255-266). New Zealand white male rabbits were allowed food and water ad libitum. Animals were fed either standard chow or 35 chow supplemented with 2% cholesterol by weight (Purina) for 10 weeks. One group of animals were injected s.c. once a day with 0.9% sterile saline and the other group of cholesterol-fed animals were administered drug (AF267B; 1 mg/kg, s.c. body weight). Following 10 weeks of treatment animals were sacrificed and evaluated for AP3 immunohistochemistry, when all sections were stained simultaneously.
WO 03/092580 PCT/L03/00357 59 Limited neuronal AP3 was observed in cortex and hilus of chow fed rabbits. Among the cholesterol-fed animals injected with saline there are abundant neurons contained identifiable Ap. Such neurons were observably smaller than those occasionally encountered in a control animal. The number of neurons expressing AP immunoreactivity was reduced 25-30% in the animals administered AF267B, 5 and the intensity of the immunoreactivity was reduced approximately 50%. It was also noted that the neurons expressing A[3 after AF267B treatment were similar in size to those encountered in control brain and therefore larger than those found in cholesterol-fed saline injected rabbit brain. These results show that AF267B is effective in decreasing elevated AP3 immunoreactivity in the brain following hypercholesterolemia, and has a neuroprotectve effect on the neurons that contain these 10 AP3 peptides. EXAMPLE 36 AF267B decreases elevated 3-amyloids in cortex in hypocholinervic rabbits (lesioned rabbits) It is known that experimentally-induced cortical cholinergic denervation results in biochemical 15 elevations of cortical AP3 concentrations and in histologic A3 deposition (Beach et al, Neurosci Lett 283: 9-12, 2000), and that administration of muscarinic Ml-selective agents to normal animals decreases CSF A3 concentrations {Beach et al Brain Res. 905: 220-223, 2001}. In the present example, animals with nbm lesions were treated with AF267B, an Mi-selective agonist, to determine whether the lesion-induced increases in CSF and cortical AP could be prevented or reduced by chronic MI receptor 20 activation. Twenty-eight female New Zealand White rabbits, about 2.5 kg each (young adults) were used. Twenty-one received lesions of the cholinergic nucleus basalis magnocellularis (nbm). The lesion was accomplished with unilateral i.c.v. injections of an immunotoxin consisting of the ribosomal toxin saporin conjugated to the monoclonal antibody ME20.4, which is directed against the low-affinity 25 neurotrophin receptor, p7 5 . The ME20.4 antibody is made against monkey p75 and also recognizes rabbit p75. The dose of immunotoxin was 32.4 ptg in 12 pl; this was delivered to the right lateral ventricle 2 mm lateral to the bregma. Seven animals received i.c.v. injections of sterile normal saline (sham lesion). Animals which received the immunotoxin were divided into 3 groups of 7. One group received twice-daily s.c. injections of AF267B; each dose was 1 mg/kg for a total daily dose of 2 30 mg/kg. Another group received physostigmine hemisalicylate in normal saline by s.c. osmotic pump at a daily dose of 3 mg/kg. The third group received twice-daily sterile saline s.c. injections. The animals which received a sham lesion were implanted with s.c. osmotic pumps filled with sterile normal saline. Animals were euthanized 4 weeks after surgery. In the case of animals receiving AF267B injections, all animals received a final injection approximately 2-3 hours before sacrifice. Four animals died 35 prematurely [(1 control animal and 3 physostigmine-treated animals), 1 to post-op hemorrhage, 1 was euthanized after developing uncontrollable seizures, 1 was euthanized due to hindlimb paralysis induced by i.m. injection of anesthetic agents prior to surgery and 1 was found dead with no cause of death found at autopsy)] and were excluded from analysis. Cerebrospinal fluid was taken from the cisterna magna of all animals at the time of sacrifice; the brain was removed and sliced coronally into WO 03/092580 PCT/IL03/00357 60 0.5 cm slices. One slice, at the level of the hypothalamus (this slice has hippocampus, as well as cortex), was fixed in 4% paraformaldehyde and processed for immunohistochemical staining with an antibody to A3. The other slices were frozen on sheets of dry ice (the other slices are the non-fixed 0.5 cm coronal slices of cerebrum, brainstem and cerebellum). Western blot analysis for A3 and sAPPce 5 was performed on the CSF from 2 of the 4 groups, those with nbm lesion and normal saline treatment and those with nbm lesion and AF267B. Quantification of the 4 kDa band representing CSF A3 showed a noticeable decrease in CSF Ap in the AF267B-treated animals versus the control animals (p = 0.05, unpaired, two-tailed t-test). There was no significant difference in the intensity of the bands representing sAPPa. Sections from the same 10 2 groups of animals stained immtunohistochemically for A3 revealed vascular A3 deposition as well as perivascular diffuse deposits in all animals. The lesion-and-treatment study showed that both AF267B and physostigmine reduced histologic deposition and biochemical levels of A3. Histologic A3 deposition was reduced to 6.4% and 12% of the lesioned, untreated group for physostigmine and AF267B, respectively. Analysis of variance found that the two treatment groups differed significantly 15 (p = .01) with respect to AD deposition (A3 deposition was high in the untreated lesioned animals vs low in the animals which were lesioned and treated with AB267B) and that both AF267B and physostigmine-treated groups differed significantly from the lesioned, untreated group (p < .05, Fisher's LSD) with respect to AB deposition (AP3 deposition was high the untreated lesioned animals vs low in the lesioned animals treated with AB267B or physostigmine). 20 The results show that AF267B treatment of animals with nbm lesions reduces the increases in CSF A3 and brain AP3 deposition that are induced by the lesion, and indicates thatMl muscarinic agonists such as AF267B may used as preventative therapy for AD. EXAMPLE 37 25 Prevention of cytotoxicity and rogrammed cell death (apoptosis) induced by various insults (deprivation from growth factors or growth factor found in serum. -amloids. oxidative stress) Confluent rat pheochromocytoma cells transfected with the M1 mAChR cultures were detached with trypsin, washed and plated in 24-well, 6-well, 60-mm or 100-mm plates that were precoated with rat tail collagen (Sigma, Israel). Several experiments were performed in serum-free medium. For 30 3-(4,5-dimethylthiazol-2-yl)- 2,5-diphenyl- tetrazolium bromide (MTT) assay, cells (1.5x104) were plated in 96-well collagen-precoated plates, in serum-free medium, with or without various drugs for 24 hours. For differentiation, cells were grown in the presence of 1% FCS and 1% HS, with addition of 50 ng/ml NGF for 7 days, to cause full differentiation. Cells were grown either on 100-mm plates (5x105 cells per plate; MTT, Fluorescence activated cell sorter (FACS) activities), or on Chamber-Slides 35 (1.5x10 4 cells; TUNEL) that were precoated with collagen, or on 13-mm glass coverslip pretreated with Poly-L-Lysine, in 24-well plates (7500 cells per well; DAPI). After 7 days, cells were washed and the medium was either replaced to serum-free, or cells were detached and replated in serum-free medium. Cells in serum-free medium were treated either with A3 peptides that were previously "aged" or with WO 03/092580 PCT/IL03/00357 61
H
2 0 2 . Tested compounds were added together with the insults for the indicated time, unless otherwise stated. Cell viability assay: 5 Cells were plated in 96-well plates in 100 pl medium. After exposure to various treatments, 10 Il of 5 mg/ml MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide; Sigma, Israel] solution in phosphate-buffered saline (PBS) was added to each well. Plates were incubated for 2 hrs at 37 0 C followed by hydrocholoric acid-isopropanol addition (100 p1 of 0.04N HC1/isopropanol). Plates were read using ELISA reader at a wavelength of 570 nm. 10 Nuclear staining of DNA: Cells were grown and treated on glass coverslips. After treatments, cells were fixed in cold methanol for 5 min at -20'C followed by treatment with cold acetone at 4 0 C for 2 min and washing in PBS. The coverslips were fluorescently stained with DAPI (4,6-diamidino-2-phenylindole; 5 mg/ml, 15 Sigma, Israel) an intercalating agent that enables visualization of chromatin condensation in the cell nuclei, for 15 min at room temperature. Cells were washed three times with PBS, mounted in a solution of glycerol containing 22 mM 1,4-diazabicyclo(2,2,2) octane (Sigma, Israel) to prevent fading, and viewed for nuclear chromatin morphology with a fluorescence microscope. Apoptotic and viable cells were counted (200 cells per coverslip, each experiment was performed in duplicates). 20 TUNEL assay: This method reveals DNA fragmentation in individual apoptotic cells. The TUNEL (Terminal deoxynucleotidyl transferase (TdT) mediated dUTP nick end labeling) method enzymatically labels DNA fragments at the 3'OH ends (representing the DNA strand breaks) with fluorescein-dUTP 25 (Boehringer Mannheim, Germany). Briefly, following treatment, cells grown on Chamber slides were fixed with paraformaldehyde solution (4%) in PBS, pH 7.4 for 30 min at room temperature. Following washing with PBS cells were permeabilized using 0.1 % Triton X-100, 0.1 % sodium citrate solution, for 2 min on ice (4 0 C), washed with PBS and 50 1 of TUNEL reaction mixture was added on each sample for 1 hr at 37 0 C, in a humidified chamber. Evans Blue reagent (diluted 1:2000 in PBS) was 30 added for 5 min and the slides were viewed with fluorescence microscope. Fluorescence activated cell sorter (FACS) analysis: Cells were differentiated for 7 days and detached from the plates with Trypsin. 106 cells were replated in 50-mm collagen-coated plates in serum-free medium in presence or absence of various 35 treatments. 200-g-centrifuiged pellets were prepared and resuspended in 300 pl PBS. 4 ml of cold methanol (-20 0 C) were added to each test tube and the fixation was carried out for 15 min at -20 0 C. Cells were washed in PBS, spun and resuspended in 1 ml PBS. Five microliters of Propidium iodide stock solution (Sigma; 10mg/ml) and 5 4l of 20 mg/ml solution of RNAse A were added for 5-10 min at room temperature. Fluorescence of individual nuclei was measured using Fluorescence Activated WO 03/092580 PCT/IL03/00357 62 Cell Sorter (FACScan; Becton Dickinson Corp.) excited at 488 nm wavelength and collected through 575±21 nm BP filter. The data were analyzed by Cell Quest software computer system. By this method we were able to measure DNA content of the cells (apoptotic cells have less DNA).Cells in G1 phase of cell cycle are after mitosis and have less DNA than cells in G2/M phase (before and during mitosis). 5 Apoptotic cells were identified as the pre-G1 phase. The MTT assay measures primary early changes within the cells, reflecting the integrity of the electron transport chain and provides a readout of cellular redox activity. This test is a specific, early indicator of the mechanism of P3-amyloid-mediated cell death and can be used to detect rapid inhibitory response. Starved, undifferentiated, rat pheochromocytoma cells transfected with MI mAChR cells L0 alone reduced cell viability by 10-20% and this effect was further augmented (up to 50-60% inhibition) by increasing concentrations of the neurotoxic full-length P3-amyloid (P3-A 1
.
42 ) peptide and its fragment, (P3-A 2 5-3 5 ) (0.5-20 .tM). When such cells were serum-deprived and treated with 3-amyloids, in the absence or presence of muscarinic agonists, the cell death induced by P3-A 25 _3s (1 pLM) after 24 hrs was significantly attenuated by addition of carbachol or AF292, AF150(S) and AF267B. 15 The potential of carbachol, AF150(S), AF267B and AF292 in protecting cells transfected with M1 mAChR cells from a direct oxidative stress induced by H 2 0 2 was tested. These agonists were observed to protect the cells from H 2 0 2 -induced toxicity. Surprisingly, muscarinic agonists were detected in compounds that have a muscarinic pharmacophore to which an antioxidant moiety is attached. These include AF604, AF700 and AF704, 20 the structures of which include a selective M1 agonist moiety linked to an antioxidant moiety. Notably, AF700, AF703 and in particular AF704 and AF704B are more effective than carbachol and AF267B against P3-A 25
-
35 (10 & 20 pM)-induced cytotoxicity. Using DAPI, an intercalating agent which enables visualization of chromatin condensation in the cell nuclei, apoptotic cell death after AP3 or H102 treatment was followed. AP3-treated cells showed the 25 morphology of apoptotic cells that shrank and lost their processes. DAPI staining revealed nuclear condensation and fragmented chromatin that is indicative of apoptotic processes. AF150(S) and AF267B were found to protect these cells from apoptosis and the neuritic-like processes are well observed. A two-fold increase in apoptotic cell death was observed after starvation for 24 hrs of the cells 30 transfected with the M1 receptor (but not in the untransfected cells). AF150(S) and AF267B significantly protect the apoptotic cell death induced by starvation. Atropine (a non-selective muscarinic antagonist) and pirenzepine (an M1 selective antagonist), reversed the protecting effect of the muscarinic agonists only in the Ml transfected cells. Treatment of cells transfected with M1 mAChr with P3-A 25 .s 35 (25 pRM) or P3-A 14 2 (25 ptM) further 35 increased by 1.5-2 fold the apoptotic cell death over starvation. The selective toxic effect of both peptides was shown using the reversed peptide (P3-A35-25) that did not induce apoptosis. AP3-induced apoptosis was prevented by AFl50(S) and AF267B only in the M1 transfected cells, while muscarinic antagonists reversed these effects.
WO 03/092580 PCT/IL03/00357 63 Oxidative insult induced by H 2 0 2 (25 and 50 pM) increased the apoptotic population by 1.5 and 2.5 fold over starvation, respectively. AF150(S) and AF267B prevented H 2 0 2 -induced apoptosis and their effect was selective to Ml mAChR activation, as atropine reversed the effect. The TUNEL method also reveals DNA fragmentation that occurs following apoptosis in 5 individual apoptotic cells. The number of TUNEL-positive stained cells increased after 25 LM P3-amyloid 25-35 treatment indicating on apoptotic process, while AF150(S) (100pM) was able to block the apoptosis so that most of the cells were TUNEL-negative stained. The cells retained their processes, and only the cytoplasm was stained in red. Quantification of apoptotic population was performed by measurement of cell DNA content, after 0 various treatments using FACS analysis. DNA histograms obtained from serum-deprived neuronal cultures in the M1 transfected cells revealed the appearance of apoptotic cell population with degraded (subdiploid) DNA content (Ml= pre-G1 phase). About 20% of total cells underwent apoptotic death after starvation for 24 hrs. Carbachol, AF150(S) and AF267B, protected cells from apoptotic death during starvation. P3-Amyloid 25-35 or 3-amyloid 1-42 increased the apoptotic population to 30-35% of 15 the cells. Co-addition of carbachol, AF150(S) or AF267B reduced the apoptotic population significantly, even below the values observed after starvation. The effect of the muscarinic agonists was blocked by 10 pM atropine indicating the involvement of M1 mAChR activation in the survival effects. Moreover, in non-transfected cells, the agonists were ineffective on starvation- and 3-amyloid-induced apoptosis. 20 EXAMPLE 38 Protection against cell death induced by N-methyl-D-Aspartate (NMDA) Primary rat brain cell cultures derived from 18-19 day old embryonic (Sprague Dawly rats) rat brains were prepared by mechano-dissociation. The brains were removed into cold Gey's Balanced salt 25 Solution (GBSS, Gibco, BRL) containing 6 mg/ml glucose. The hippocamous and cortex were separated and transferred to Dulbecco's Modified Eagle Medium (DMEM, Biological Industries, Bet-Haemek, Israel) containing 6 mg/ml glucose, 2 mM L -glutamine (Biological Industries, Bet-Haemek, Israel), 1000 I.U/ml Penicillin G sodium and 3% ultrosere G (Gibco, BRL). Following cell dissociation using a fire polished Pasteur pipette, the resulting cell suspension was plated on tissue 30 cultures precoated with poly-L-lysine (30,000-70,000 MW, Sigma) 1 mg/ml in borate buffer. Cells were plated at a density of 80000 cells/well on a 96 well culture plate, or at 400000 cells/well (hippocampal cells) and 600000 cell/well (cortcal cells) on 12 well culture plates. Cell cultures were maintained in growth cell medium at 37 0 C 5%CO2/95% 02 for about 2 weeks. Glial cell proliferation was arrested following 3-4 days in culture by addition of 5-fluoro-2'-deoxyuridine/uridine/cytosine 35 arabinoside mixture (5 mM final concentration). For evaluating exposure to NMDA, cortical and hippocampal cell culture at 10 days in culture plated in 96 well culture plates were exposed to 100 or 200pM NMDA (RBI, USA). Neuroprotective potency of compounds from formula I was compared to that elicited by 20M of MK801 (a non-competitive NMDA receptor antagonist). Cell cultures were also exposed in parallel to medium WO 03/092580 PCT/IL03/00357 64 alone and were referred as to controls. In order to produce widespread neuronal injury, all exposures were carried out for 20-24 hrs prior assessing neuronal cell death. Neurotoxicity was quantitatively assessed by measuring the extent of mitochondrial activity in living cells using {2,3-bis[2-methoxy 4-nitro- 5-sulphopheny]- 2H-tetrazolium- carboxanilide inner salt]}-based assay (Klausner, Biotechnol 5 5:779-786, 1987; Lipman et al, Cytotechnol 8:129-176, 1992). AF150(S) and AF267B were effective in prevention of cell death. The neuroprotective potency of 100gM of these agonists against 100pM NMDA-induced toxicity was similar to that elicited by 20pM MK801, but slightly less against 200gM of NMDA. [0 EXAMPLE 39 1. Effects on tau protein hyperphosphorylation: Primary cell cultures were grown as described in EXAMPLE 38. Cell supernatant were removed and the cells were washed once in medium prior to the addition of cold phosphate buffre saline ph=7.4 (PBS) solution containing 0.2 mM EDTA. Cells were scraped using a rubber policeman, transferred to 15 Eppendorf tubes, and centrifuged at 4 0 C. Cell pellets were resuspended in lysis buffer (EDTA 5 mM, Tris 50 mM, Triton 1%, NaC1 150mM) containing protease inhibitors (5 units/ml apronitin, 5 mg/ml pepstatin, 5mg/ml leupeptin and 0.1mM PMSF, Sigma) and subjected to centrifugation at 40C. Supernatants were transferred into Eppendorf tubes and kept at -200C till analyzed. The extent of tau-1 immunoreactivity (an antibody that recognizes a non-phosphorylated tau at Ser 199 ) was determined by 20 western blots. AF150(S) and AF267B were effective in elevating tau immunoreactivity both in cortical and hippocampal cell cultures in a dose range of 1-100gM. 2. Effects on tau phosphorylation and antagonism of Ap-induced effects on tan phosphorviation 25 Following the experimental design of Sadot et al., J.Neurochem. 66:877-880, 1996, including immunobloting with AT8 antibody, an antibody that recognizes phosphorylated tau at Ser 199
/
202 , it was found that AF267B, AF292 AF704, and AF704B (100 tM) induce dephosphorylation of tau proteins in these cell cultures to the control level. By linking an Ml agonist to an antioxidant moiety tau phophorylation may be decreased, providing a new therapeutic strategy in a variety of CNS disease 30 states due to combined damage due to oxidative stress and tau hyperphosphorylation. EXAMPLE 40 Effects on ApoE synthesis and secretion in rat primary type 1 astrocyte cultures Primary cultures of type 1 astrocytes were derived from the cortex of Sprague Dawley rats. The 35 compounds tested were dissolved in the culture media and added to cells for 24, 48, 72 and 96 hrs. Media was then removed and kept frozen until ApoE protein levels were evaluated by immunoblot analysis as described in Poirier et al Neuroscience 55: 81-90 (1993). Alternatively the methods of Cedazo-Minuez et al [Neurosci 105: 651-661, 2001} may be employed. AF102B at 96 hrs was inactive toward ApoE metabolism (synthesis and secretion). The compound AF267B, and to a lesser WO 03/092580 PCT/IL03/00357 65 extent AF150(S), inhibits the production of ApoE over time with a maximal effect at 96 hrs. The effects observed (60-80% inhibition) occur at 0.1 uM, a very low concentration of the compound. These results indicate that this compound inhibits apoE production, including ApoE4 in rat astrocytes, and thus may be used in therapies in which inhibition of ApoE production is indicated. 5 EXAMPLE 41 Competition binding assay for muscarinic receptor with an agonist as the labeled probe. Oxotremorine-M (OXO-M) is an agonist that binds to all muscarinic receptor subtypes with similar affinities. The ability of a test compound to displace [ 3 lH]OXO-M binding provides a measure 0 for the affinity of the test compound to the receptor agonist binding site. The competition of
[
3 H]OXO-M binding with AF292 and its enantiomer, AF291, have Ki values of 0.27 and 6.56 jiM respectively as compared to the full agonist carbachol which shows a Ki of 0.05 iM. Pirenzepine (PZ), a muscarinic antagonist, binds preferentially to M1 receptors while OXO-M binds to all mAChR subtypes non-selectively; the ratio between the Ki values for PZ versus that of S5 OXO-M may be indicative of the selectivity of the tested compound. The smaller the ratio, the more MI selective is the tested compound. Competition of [ 3 H]PZ binding to rat cortical membranes with AF292 (Ki = 1.39 pM) and its enantiomer AF291 (Ki = 10.7 jiM) shows that AF292 is the active enantiomer. The OXO-M/PZ ratio is -0.36 showing a moderately high selectivity for the M1 receptor. 20 EXAMPLE 42 Muscarinic receptor selectivity 1. Functional studies in cell cultures transfected with human muscarinic receptor subtypes 1.1 AF292 was tested for its agonistic or antagonistic properties, its potency, and its selectivity towards the human M1 vs. M3 and M5 receptors in activating phosphoinositide (PI) hydrolysis 25 according to the method of Gurwitz et at Eur. J. Pharmacol. 267, 21, 1993. The effects of the compounds were atropine-sensitive in activating PI hydrolysis demonstrating, their muscarinic nature. AF292 and AF267B were found to be partial agonists at the M1 receptor, showing -35% and -66% activity, respectively, versus carbachol with respect to PI turnover measured in this paradigm. No activity was seen at the M3 and MS receptor with AF292, as compared with AF267B (-30% vs. 30 carbachol at the M3). AF292 was both a partial agonist at the M1 receptor and a weak antagonist at the M3 receptor (pKb = 0.66 pM), with no agonistic activity at M2 or M5 mAChR. The effects on the M2 receptor were measured in modified cell cultures that show an increase in intracellular Ca ions following activation with carbachol. In spite of an 8-fold increase in activity induced by carbachol on the M2 mAChR, AF292 was inactive as an agonist at all tested concentrations (10 " 9 -10- 3 M). 35 1.2. AF292 was tested for its agonistic or antagonistic properties, its potency, and its selectivity towards the human Ml vs. M3 and M5 receptors in activating arachidonic acid (AA) hydrolysis according to the method of Gurwitz et al Eur. J. Pharmacol. 267, 21, 1993. AF292 was more potent on AA release induced by Ml mAChR (80%) than on PI turnover (35%), but still inactive as an agonist WO 03/092580 PCT/IL03/00357 66 (AA release) on M3 and M5 mAChR. Thus AF292 is a more efficacious agonist on M1 mAChR-mediated AA release (mediated via phospholipase A2) than on PI (mediated via phospholipase C). In summary, not only is AF292 highly selective for the M1 mAChR as an agonist, but it also exhibits distinct activation of select G-proteins (e.g. not all the G-proteins are activated to the same 5 extent by AF292, unlike carbachol, which acts as a non-selective mAChR agonist that activates all these receptors to the same extent. 2. Binding studies to muscarinic receptors and other systems 2.1 In competitive binding studies against the following ligands for mAChR receptors, AF267B was 10 found to be highly selective for the following M 1 mAChR subtypes: QNB [Muscarinic Antagonist in Rat Cortical (CTX) Membranes] (Ki= 49.6±9 pM); QNB [Muscarinic Antagonist in Rat Cerebellar Membranes] (Ki = 45.2±10.8 RM); Pirenzepine (M 1 selective Antagonist in CTX) (Ki = 3.74±0.59 pM); Oxotremorine-M (Muscarinic Agonist in CTX) (Ki = 1.62±0.34 pM)]; vs. Serotonin, 5HT3 51.3% inhibition at 10 4 M; Opiods/Opiate, Non-Selective 52.5% inhibition at 10 4 M; with no binding at all to 15 Adrenergic(A), alA, Adrenergic; alB, Adrenergic; c2A (Human Recombinant); Adrenergic, cx2B; Adrenergic, a2C (Human Recombinant); Adrenergic, P31; Adrenergic, 12; Benzodiazepine (BZD), Peripheral; Clozapine; Dopamine, Dl; Dopamine, D2 (Human Recombinant); Dopamine, D3 (Rat Recombinant); GABA A, Agonist Site; GABA A, Benzodiazepine, Central; Glutamate, AMPA Site; Glutamate, Kainate Site; Glutamate, NMDA Agonist Site; Glutamate, NMDA, Glycine; Glycine, 20 Strychnine-Sensitive; Histamine, H1; Histamine, H2; Histamine, H3; Nicotinic; Ganglionic site; Nicotinic, Neuronal site; Serotonin, 5HTIA (Human Recombinant); Serotonin, 5HTIB; Serotonin, 5HT 4 : Serotonin, 5HT 6 (Rat Recombinant); Serotonin, 5HT 7 (Rat Recombinant); Choline Acetyltransferase; Glutamic Acid Decarboxylase; Monoamine Oxides A, MAO-A; Monamine Oxidase B, MAO-B. 25 2.2. In binding studies, AF292 (10 pM) was found to be highly selective for the mAChR subtypes {M1 (human) (55%), M2 (human) (61%), M3 (human) (55%)} with no binding at all to: adenosine Al (human); A2A (human); adenosine A3 (human); alpha 1 adrenergic (non-selective); alpha 2 (non-selective); beta 1 (human); angiotensin, AT1 (human recombinant); benzodiazepine (BZD) 30 (central); bradykinin, B2 (human recombinant); cholecystokinin (CCKA) (human recombinant) (CCK1); dopamine D1 (human recombinant); D2S (human recombinant); endothelin, ETA (human recombinant); GABA (non-selective); galanin, GAL2 (human); chemokine, IL-8B (human recombinant) (CXCR2); chemokine, CCR1 (human recombinant); histamine, H1 (central); histamine, H2: melanocortine, MC4 (human recombinant); melatonin, ML1; tachykinin, NK2 (human 35 recombinant); NK3 (human recombinant); neuropeptide, Y1 (human); neuropeptide, Y2 (human); neurotensin, NT1 (human recombinant) (NTS1); opiate, delta 2 (human recombinant) (DOP): opiate, kappa (KOP); opiate mu (human recombinant) (MOP); orphanin, ORL1 (human recombinant) (NOP); serotonin, 5-HTIA (human recombinant); serotonin, 5-IHTiB; serotonin, 5-HT2A (human recombinant);serotonin, 5-HT 3 (human recombinant); serotonin, 5-HT5A (human recombinant) WO 03/092580 PCT/IL03/00357 67 (5-ht5A); 5-HT 6 (human recombinant); 5-HT 7 (human); somatostatin, sst (non-selective); vasoactive intestinal peptide, VIP1 (human) (VPAC1); vasopressin Vla (human recombinant); Ca 2+ channel (L, verapamil site); K+V channel; SK+Ca channel; Na + channel (site 2); C- channel; norepinephrine NE transporter (human). 5 EXAMPLE 43 Effects in aging microcebes Aging microcebes show similar cognitive deficits and cerebral lesions to those observed in aging humans and in AD patients. Thus this is a good animal model for AD, mimicing the three major 10 hallmarks in AD {plaques (AP3), paired helical filaments (hyperphophorylated and aggregated r) and cognitive dysfunction}. This model may also be used to mimic MCI conducive to AD In this model AF150(S) [chronic treatment for 18 months]: i) improved the cognitive and behavioral impairments ii) decreased hyperphosphorylated c proteins and the number of neurons containing aggregated - protein (e.g. indicative of diseased brains) and the number of paired helical 15 filaments; and iii) decreased astrogliosis and inflammation. This indicates AF150(S) may be used as a drug to treat or modify the effects of AD but does not produce tolerance following prolonged treatment. EXAMPLE 44: M1 agonists reduce neurobehavioral impairments following closed head injury in mice 20 Closed head injury (CHI) was induced in mice as described in Chen et al, J Neurotrauma, 15: 231-237. 1998. Neurological severity scores (NSS) were assessed using a battery of 10 parameters (10 = worst outcome, 0 = normal fimunction). The compounds tested (1 mg compound/kg body weight) vs. placebo-treated animals were injected ip 5 min after CHI vs. placebo-treated animals. Treated mice were evaluated at 1 h, to determine the severity of injury, and at 24 and 48 h to determine recovery. The 25 NSS was as follows: 1. Control (N=10): 7.80+/-0.25 (lh); 5.30+/-0.33 (24h); 4.20+/-0.47 (48h). 2. AF150(S) (N=10): 8.00+/-0.21 (lh); 4.30+/-0.26**(24h); 2.90+/-0.
23 b (48h); AF267B (N=9): 7.89+/-0.26 (lh); 3.67+/-0.24* (24h); 2.89+/- 0.260 (48h) [*p=0.0 3 ; **p-0.03; ap =0.009; bp=0.005; op=0.004]. All the compounds tested showed a highly significant improvement on the motor functions. 30 Recovery was faster in the AF267B treated animals in two balance tests (beam walk) - [22% vs 80% in control at 24 h (3cm) or 33% vs. 80% in control at 48 h (2cm)]. EXAMPLE 45: AF150(S). AF267B in social memory in rats 35 Social olfactory recognition in rodents has been shown to assess short term memory and to be sensitive to cholinergic drugs (Dantzer et al. Psychopharmacol. 91:363-368, 1987; Perio et al Psychopharmacol. 97: 262-268. 1989). In this example the effect of AF150(S) and AF267B on investigatory behavior of naive rats was tested.
WO 03/092580 PCT/IL03/00357 68 12 Male Wistar rats, 400-530 gr (4-5 months old) were used. Rats were housed individually 14 days before testing. Juvenile Wistar rats 40-50 gr (at arrival) were kept in groups of 6 and served as social stimuli for the adult rats. Animals were kept at 210 C_1, with an inverse light- dark cycle (light on from 2:00 P.M. to 2:00 A.M.). The sessions were conducted 7 hr into the dark part of the light/dark 5 cycle, under red illumination. Adult rats were placed in a dim illumination room 1.5 h before the beginning of the social test. All juveniles were isolated in cages for 30 min prior to the beginning of the experiment. At the beginning of testing, an unfamiliar juvenile rat was placed in the home cage of an adult rat for 5 min. The time spent by the adult rat in investigating the juvenile rat was recorded. The adult rat was then 10 immediately (1-2 min) treated with vehicle or test compound. Two hours later, the same juvenile was presented to the same adult rat for another 5 min period, a time when normally the stimulus juvenile is no longer identified (i.e., the adult rat investigates the juvenile for the same amount of time as during the first presentation). Thus under the influence of a purported memory enhancing drug, the time spent in investigating the same juvenile is expected to be reduced. Two days later, a juvenile, different from 15 the one used for the first exposure, was presented to the adult rat, 2 h after drug or vehicle administration. Any reduction in social exploration of this different juvenile is thus considered as reflecting a nonspecific effect of the drug (i.e., not memory related). On no occasion was a subject tested twice with the same juvenile stimulus animal, nor was a juvenile used more than once in a 48-hr period. 20 AFl50(S) or AF267B (0.5, 1 and 5 mg/kg, p.o.) or vehicle, phosphate buffer saline (PBS) were administered to the adult rats immediately after the first exposure to the juvenile rat. Time spent in social investigation of stimulus juvenile rat was measured (in sec) and then expressed for each animal as the ratio of the second exposure to the first exposure (Ratio of Investigation Duration (RID)). This transformation to RIDs was used in order to minimize possible 25 individual as well as day-to-day variations in baseline performance (Perio et al Psychopharmacol. 97: 262-268. 1989). Therefore any reduction in investigation time during the second exposure will lead to a RID which is less than 1, indicating that the animal recognizes the juvenile rat. Analysis for repeated measurement was made by a 3-way ANOVA and post hoc comparisons were made by simple main effects contrasts analysis. 30 AFl50(S) and AF267B decreased the investigation time of the same juvenile compared to placebo group in a dose-dependent manner. This improvement of memory cannot be attributed to non-specific effects, since it was not observed when a different juvenile was used for the second exposure. Both compounds thus appear to facilitate social memory in naive rats. No significant difference was found between the total RIDs of the two compounds, but the 35 interaction between similar/ different juvenile x doses of both drugs was found statistically significant, [F(3/33)= 14.9, p< 0 .0001]. Specifically, both compounds significantly reduced the investigatory time of the same juvenile at all three doses tested (p<0.001) relative to placebo. Furthermore, a significant difference was found between the RIDs of the 0.5mg/kg and the two other doses (p<0.05); The RIDs observed for the 1 and 5 mg/kg were significantly lower than those observed for the 0.5mg/kg.
WO 03/092580 PCT/IL03/00357 69 It should be noted that a significant difference was found between the RIDs of the same juvenile group and a different juvenile group at all three doses (p<0.01- p<0.001). EXAMPLE 46 5 Effects of AF267B and AF292 on passive avoidance (PA) in cholinotoxin (AF64A)-treated rats The general procedure followed is described in Fisher et al, J. Pharmacol. Exptl. Therap., 257: 392, 1991. AF64A (10 mM) was prepared by alkaline hydrolysis of acetylethylcholine mustard.HC1. Rats, anesthesized with Equithesin (0.3 ml/100 g, IP) were injected bilaterally by stereotaxic application of AF64A (3 nmol/2 1 .l/side) or saline (2 pl) into the lateral cerebral ventricles (AP = -0.8; 10 L = ±1.5 mm from bregma; and DV = -4.8 mm from skull surface). Infusions were made via a CMA 100 microinjection pump, through a 30-gauge injection carmnnula, at a constant rate of 0.25 gl/min. The cannula was left in place for 4 min after injection to allow diffusion of the solution into the ventricles. Compounds or phosphate-buffer-saline (PBS) were administrated once, p.o., immediately after shock. Retention was tested 72h after training. 15 A significant difference was found in the initial latency between all AF64A-injected rats (29.02±3.1s) and all saline injected rats (20.55±1.95s), F(1/72)=5.13, p<0.05. A statistically significant interaction was found between AF64A-injection x drug treatment, F(3/72)=11.99, p<0.001, in the retention latency. The retention latency of AF64A-injected rats treated with PBS (67.1±18.9s) was significantly shorter (poorer memory) than that of saline-injected rats treated with PBS (455.3±55.1) 20 (p<0.001, by simple main effects contrasts analysis). The retention latencies of AF64A-injected rats treated with AF267B, 0.1mg/kg (440.7±46.4s), and AF292, lmg/kg (447±46.8s), were significantly longer (better memory) than that of AF64A-injected rats treated with PBS (p<0.001, by simple main effects contrasts analysis). No significant difference was found between the retention latency of AF64A-rats treated with AF267B, 25 0.03mg/kg (105.7±31.9s), and that of AF64A-rats treated with PBS. No significant differences were found between the latencies of any of the saline-injected groups. AF64A-injected rats demonstrated a clear impairment in retention of the PA task. The minimal effective dose of AF267B in attenuating AF64A-induced retention deficiencies is less than 0.1mg/kg, p.o. Both AF267B, 0.1mg/kg, and AF292, 1mg/kg, in the PA task, are efficacious in improving 30 AF64A-induced retention deficiencies, compared to AF64A-injected rats treated with PBS. The minimal effective dose of AF292 may be below 1 mg/kg, po. EXAMPLE 47 Effects of AF267B cognitive impairments induced by AF64A in rats in the MWM test. 35 AF64A or saline-injected (6 months old) Sprague-Dawley rats were tested in the Morris Water Maze (MWM) task. The paradigm used assesses spatial learning abilities in a reference memory regimen, and involves training (days 1-4), transfer test (Probe trial - day 4, 3 min following the last training trial) and reversal test (day 5).
WO 03/092580 PCT/IL03/00357 70 At 4 months post-operation, each of the AF64A and saline groups of rats was randomly subdivided into four treatment subgroups (n=9): subgroups 1 - 3 were treated with AF267B in doses of 0.3, 1, and 3 mg/kg, po, in a volume of 10 ml/kg, whereas subgroup 4 (control group) was treated with the vehicle, phosphate-buffer saline, 10 mM (PBS) in the same volume. Drugs and PBS were 5 administered once a day for 5 days before starting the behavioral testing, and then for the duration of the 5-day experiment, 30 min before testing. The three measures, escape latency, path length and swimming speed were analyzed by MANOVA, followed by simple main effects contrast analysis. Results: AF64A-injected rats showed a significantly longer escape latency than saline-injected rats, 10 F(1/64)=10.56, p<0.005. In terms of path length, AF64A-injected rats showed a significantly slower learning curve than saline-injected rats, F(3/192)=4.01, p<0.01. AF267B had no significant effect on learning; however, AF64A-injected rats treated with AF267B-1 mg/kg showed a tendency for improvement, in escape latency only, while AF267B-3 mg/kg tended to impair performance in these rats. No correlation was found between the cognitive measures (escape latency and path length) and the 1 5 nonspecific, motor measure (swimming speed). All saline-injected rats showed a spatial bias in the probe trial, in both parameters, F(3/192)=7.86, p<0.001, and F(3/192)=7.44, p<0.001, for escape latency and path length, respectively. On the other hand, AF64A-injected rats treated with PBS showed only a partial spatial bias on this test. However, AF64A-injected rats treated with AF267B showed a complete spatial bias, similar to that of 20 saline-injected rats, as presented in escape latency only, F(9/192)=2.3, p<0.025. No significant differences were found between the various doses of AF267B, in their beneficial effect on memory. No significant differences were found between any of the groups tested in the reversal test. However, AF64A injection tended to deteriorate cognitive performance, in both measures. Additionally, AF64A-injected rats treated with AF267B-1 mg/kg showed a tendency for improvement, 25 while AF64A-injected rats treated with AF267B-3 mg/kg showed a tendency for impairment on this test. EXAMPLE 48 The effects of AF150(S, AF267B. rivastiamine and nicotine on MWM performance of C57BL/10SnJ 30 vs. C57BL6J mice C57BL/10SnJ (B 10) mice were selected due to their small hippocampi and decreased number of hippocampal pyramidal neurons; the cell loss seemed to be associated with poor spatial learning. Deficiencies in spatial memory tasks observed in these animals were reported to be responsive to cholinergic manipulation (scopolamine) (Simons et al Life Sci., 42, 375-383, 1988), and both AChE 35 inhibitors (physostigmine) and muscarinic agonists (AF102B, PD151832) (Simons et al., 1988; Vincent et al Brain Res., 597, 264-268, 1992; Schwarz et al Drug Dev. Res., 40, 133-143, 1997) have shown positive effects in this model, using the MWM. Each group of mice was randomly divided into 7 treatment groups (n=12-14/ group). Groups 1-2 were treated with AF150(S) at doses of 0.5 and 1 mg/kg, i.p, in a volume of 10ml/kg, groups 3-4 were WO 03/092580 PCT/IL03/00357 71 treated with AF267B at the same doses and volume, groups 5-6 were treated with rivastigmine and nicotine, respectively, at the dose of 1 mg/kg, i.p., and group 7 was treated with the solvent, saline 0.9%. All tested compounds and saline were administered once a day for 4 days before starting the behavioral testing, and then for the duration of the 5-day experiment, 30 minutes before testing. 5 Training: Each mouse was trained for four consecutive days, four trials (one block) per day, in which the platform position remained constant and was located in the center of the southeast quadrant of the pool. Within each block of four trials, each mouse started at each of the starting locations, but the sequence of locations was randomly selected. A trial consisted of placing a mouse by hand into the water facing the wall of the pool at one of four starting locations, north, south, east or west, around the 10 pool's perimeter. Escape latency (the time to find the platform), path length (the distance traveled by the mouse) and speed (the swimming rate of the mouse) were recorded on each trial by the monitoring system. For each mouse, the path length, escape latency, and swimming speed of the four trials on each of the 4 training days were grouped into blocks (one block for each day). The scores of all three measures 15 were analyzed by a three-way MANOVA (2 x 7 x 4) with one repeated variable (days) and two non repeated variables [mice strain - C57BL/10OSnJ or C57BL6J, and treatment - two doses of each, AF150(S) and AF267B, rivastigmine, nicotine (one dose for each) and saline]. Specific comparisons were performed, using the simple main effects contrasts analysis, which is specifically suited for testing significant interactions. 20 Escape latency. Small-hippocampi mice showed significantly longer escape latencies (indicating a worse RM performance) than normal hippocampus rats. AF150(S) and AF267B, and rivastigmine, positively affected the training performance of small hippocampus mice, F(6/161)=6.39, p<0.0001. Specifically, both doses of each of the muscarinic compounds improved the escape latencies of small hippocampus mice, compared to control group (p<0.01-p<0.001); Furthermore, AF267B showed a 25 dose-response curve in its effect on performance (p<0.02) while AF150(S) affected performance equally by both doses. Both AF150(S) and AF267B affected performance more effectively than rivastigmine (p<0.05-p<0.001, respectively). AF150(S) increased the escape latencies of normal hippocampus mice, by both doses (p<0.05-p<0.01) whereas AF267B did not significantly affect the escape latencies of these mice during training. While nicotine had no improving effect of memory 30 deficits shown by small hippocampus mice, it degraded the performance of normal hippocampus mice (p<0.02). The results also indicated a significant general effect of training, F(3/483)=90.49, p<0.0001; the escape latencies of all groups decreased linearly during the four training days (p<0.0001, by a polynomial contrast). Path length. Small hippocampi mice showed significantly longer path lengths than normal 35 hippocampus mice, F(6,161)=2.35, p<0.0 33 . AF150(S) (both doses), AF267B (the higher dose) and rivastigmine positively affected the performance of small hippocampus mice (p<0.05-p<0.01). AF150(S) (only the higher dose) significantly (p<0.05) impaired the path length of normal hippocampus mice whereas neither AF267B nor rivastigmine had any effect on the path length of these mice. Nicotine had no significant effect on the performance of any of the mice strains tested. The WO 03/092580 PCT/IL03/00357 72 results also indicated a significant general effect of training, F(3/483)=86.98, p<0.0001; the path lengths of all groups decreased linearly during the four training days (p<0.0001, by a polynomial contrast). Swimming speed. Motor activity differences were observed between the two strains of mice 5 treated with saline: The swimming speed of small hippocampus mice was significantly lower than that of normal hippocampus mice, F(6/161)=14.32, p<0.0001. Furthermore, both muscarinic drugs significantly increased the swimming speed of small hippocampus mice. Specifically, AF267B enhanced the swimming speed in a dose dependent manner (p<0.001, relative to control; p<0.02, between doses) while the enhancing effect of AF150(S) was equal in both doses (p<0.001). Moreover, 10 AF267B-rmg/kg significantly enhanced the swimming speed more strongly (p<0.001) than AF150(S)-I mg/kg. Neither rivastigmine nor nicotine had any significant effect on the swimming speed of small hippocampus mice. AF150(S) significantly (p<0.0 l-p<0.001) impaired the swimming speed of normal hippocampus mice while AF267B had no such effect. Likewise, rivastigmine (p<0.01) and nicotine (p<0.001) significantly decreased the swimming speed of these mice. The results also indicated 15 a significant general effect of training, F(3/483)=15.34, p<0.0001; the swimming speeds of all groups increased linearly during the four training days (p<0.0001), by a polinomial contrast). Transfer test. During trial No. 17, on the fourth day, the platform was entirely removed from the pool (a probe trial). In this trial, the mouse was placed into the water for a limited period (30 s), and its spatial bias was measured by recording the relative distribution of escape latency and path length over 20 the four quadrants of the pool. The path length and escape latency for the transfer trial (trial No. 17) were analyzed by a three-way MANOVA (2 x 7 x 4) with one repeated variable (quadrant in the pool) and two non repeated variables [mice strain - C57BL/10SnJ or C57BL6J, and treatment - two doses of each, AF150(S) and AF267B, rivastigmine and nicotine (one dose for each) and saline]. The three-way interaction for the escape latency measure was found statistically significant, 25 F(18/483)=1.62, p<0.05, while the interaction for the path length measure was close to significance, F(18/483)=1.5, p<0.08. Normal hippocampus mice treated with saline showed a complete spatial bias in the transfer test. They spent significantly more time in the training quadrant (p<0.001) relative to the three other quadrants of the pool. On the other hand, small hippocampus mice treated with saline showed only a partial spatial bias in this test; They spent significantly more time in quadrant No. 1 30 relative to quadrants No. 3 (p<0.001) and 4 (p<0.05) but not relative to quadrant No. 2. However, small hippocampus mice treated with AF150(S) or AF267B (by both doses), or rivastigmine, showed a complete spatial bias, like normal hippocampus mice. In contrast, small hippocampus mice treated with nicotine showed only a partial spatial bias, like small hippocampus mice treated with saline. Normal hippocampus mice treated with AFl50(S)-l mg/kg showed only a partial spatial bias in the transfer test 35 while all other normal hippocampus mice treated with the other drugs showed a complete spatial bias in this. test. The results of the path length measure were very similar to those of the escape latency measure. Reversal test. During trials 18-21, on the fifth day, the platform position was changed to the northwest quadrant, opposite to the training quadrant. Thus, during reversal learning, the platform WO 03/092580 PCT/IL03/00357 73 location was moved relative to the configuration of objects within the room, but the pool occupied the same place within the room throughout the entire experiment. Testing of the rats and measures taken were the same as in training. For each mouse, the escape latency, path length and swimming speed of the reversal test (trials 5 No. 18-21) were grouped into one block. All three measures were analyzed by a two-way MANOVA (2 x 7) with two variables ([mice strain - C57BL/10SnJ or C57BL6J, and treatment - two doses of each, AF150(S) and AF267B, rivastigmine, nicotine (one dose for each) and saline]. Escape latency. Small hippocampi mice showed significantly longer escape latencies during reversal learning than normal hippocampus mice, F(6/161)=3.26, p<0.005. Both muscarinic drugs, 10 AF150(S) and AF267B (by both doses), significantly (p<0.05-p<0.01) improved the escape latency of small hippocampus mice while AF150(S)-0.5 mg/kg significantly (p<0.05) impaired the escape latency of normal hippocampus mice. Both rivastigmine and nicotine had no significant effect on either C57BL/10 SnJ or C57BL6J mice. Path length. The only significant effect (p<0.05) shown in this measure was the impairment of 15 reversal learning by AF150(S)-lmg/kg in normal hippocampus mice [F(6/161)=2.85, p<0.011, for the two-way interaction). Swimming speed. No significant differences were obtained between the saline-treated, two strains of mice in motor activity. However, both muscarinic drugs, by both doses, significantly (p<0.01-0.001) increased the swimming speeds of small hippocampi mice, F(6/161)=8.71, p<0.0001. 20 The swimming speeds of normal hippocampi mice were significantly decreased by AF150(S)-0.5 mg/kg (p<0.02), AF267B-lmg/kg (p<0.05), rivastigmine (p<0.01) and nicotine (p<0.05). AF150(S), AF267B, and the AChE inhibitor, rivastigmine, significantly attenuated these impairments in mice with small hippocampus. The improvement of cognitive functioning was more pronounced during acquisition and retention, although a similar improvement was shown by both 25 muscarinic compounds in reversal learning. In contrast, nicotine had no beneficial effect on the cognitive performance of small hippocampi mice. A dose-response effect of AF267B was demonstrated in acquisition, by the differential improvement of the cognitive deficits shown in escape latency measure. The beneficial effect of the 1 mg/kg dose was significantly stronger than that of the 0.5 mg/kg dose. During transfer trial, non-treated, small hippocampi mice showed only partial memory deficits 30 concerning the platform location. A significant improvement of these deficits was demonstrated equally well by both muscarinic drugs, at the two doses tested, as well as by rivastigmine, but not by nicotine. The contribution of AF267B and AF150(S) to the improvement of learning and memory processes is emphasized by two findings: the dose-response effect shown by AF267B in acquisition, and the beneficial effect of both drugs demonstrated in the probe (transfer) trial. In this respect it should be 35 noted that the probe trial is the foremost procedure in the MWM task, providing measures that quantify the strength and accuracy of the original learning.
WO 03/092580 PCT/IL03/00357 74 EXAMPLE 49 AF150(S) is effective in restoration of conitive impairments in ischemic rats. Transient ischemia in rats was induced by a modification of the ischemia model (Voll et et al, Stroke, 20: 1700-1706, 1989). This was done by a bilateral carotid artery occlusion in Sprague Dawley 5 rats: (42 male, 3 months old, weighing 270-340 g) combined with reduction in blood pressure induced by sodium nitropruside. Ischemia was induced in 21 rats whereas the other 21 rats served as sham controls. Under pentobarbital anasthesia (30mg/kg, ip), sodium nitropruside 4.8mg/kg/hr) was infused through a cannula implanted in the tail vein, for a period of 25 min. Five min after the initiation of the infusion, at the time when mean blood pressure was maintained at 30-60mmHg (intial levels -110mm 10 Hg) both carotid arteries were clamped for 20 min. Immediately afterwards 1.8mEq sodium bicarbonate solution was administerd ip in order to minimize systemic acidosis. The sham operated rats were anesthetized as the ischemic rats and were infused with saline. Their carotid arteries were exposed but were not subject to carotid clamping. In rats subjected to ischemia, a mortality of about 30% was recorded within 24 hr after surgery. Animals were allowed to recover for 3 weeks prior to behavioral 15 testing. Rats were randomly assigned to one of four groups: ischemic and sham-operated rats which were treated with AF150(S) (0.5 mg/kg, po) and ischemic and sham-operated control rats treated with double distilled water (DDW) (10ml/kg, po). Each groups comprised of 10-11 rats. AF150(S) was administerd immediately following operation, once a day (6 days/week) for weeks before starting the behavioral testing, and then for the duration of the three-weeks experiment, 60 min before testing. The 20 evaluation of the animals was done using the working-memory matching-to-sample paradigm in the MWM. The ratio of escape latency (REL) and the ratio of path length (RPL) were calculated by the ratio of block no 2/block no 1 for each parameter. REL and RPL reflect the relative saving in performance from trial no 1 to trial no 2. REL and RPL were analyzed by a 3-way ANOVA (2x2x3), with one 25 repeated variable (weeks) and two non-repeated variables (Operation- ischemia/sham-operated and Treatment-AF150(S)/DDW). For both REL and RPL the interaction between operation x treatment was found statistically significant [F(1/32)=8.08; p<0.01 and F(1/32)=6.75; p<0.025, for REL and RPL, respectively). Simple main effects contrasts analysis showed that both REL and RPL of ischemic rats treated with DDW were 30 higher than those of control rats treated with DDW (p<0.01 and P<0.02 for REL and RPL, respectively). This result indicates a deficit in working memory processes of ischemic rats compared to control rats. AF 150(S) significantly improved working memory performance of ischemic rats, compared with DDW-treatment; both REL and RPL of ischemic rats treated with AF150(S) were significantly lower 35 (p<0.05) than those of ischemic rats treated with DDW. Control rats treated with AF150(S) did not show any significant change in performance. No differences in swimming speed were found in any of the tested groups. In concluson, chronic administration of AF150(S), 0.5mg/kg, po, showed a clear improvement of working memory performance in ischemic rats during the three weeks of the experiment (following 3-6 weeks of drug administration). Nonspecific, motor coordination effects WO 03/092580 PCT/IL03/00357 75 could explain neither the behavioral effects of the ischemic rats, nor the improving effects of AF150(S), because no significant effects were demonstrated in the swimmig abilty of the rats in the Morris water maze testing. 5 EXAMPLE 50 Effects in Trihexvphenidyl treated rats - AF150(S), AF267B, AF292, AF704 Trihexyphenidyl is a selective M1 muscarinic antagonist that crosses the blood brain barrier and induces memory and learning impairments (Bymaster et al. J Pharmacol Exp Ther 267: 16-24, 1993. Roldan et al Neurosci. Lett. 230: 93-96, 1997; Kimura et al Brain Res. 834: 6-12, 1999). 10 Naive Wistar rats were used in the experiments below. The passive avoidance (PA) task is comprised of training (acquisition) phase and a retention phase. In the training procedure each rat was individually placed in the small illuminated compartment and after 60 sec. of familiarization/ adaptation, the door to the large compartment was opened and the latency to enter was measured (Initial Latency). Immediately following entry into the dark compartment, the door was closed and inescapable 15 foot shock (0.6mA for 3 sec) was delivered through the grid floor. A cutoff point of 180 sec was used for initial latency. Animals that failed to enter (step-through) within 180 see were excluded from the experiment. After the acquisition trial the rat was returned to its home cage. Retention of the passive avoidance task was measured 24h later, by again placing the rat in the light compartment and after a 60 sec adaptation interval, the door was opened and the latency to re-enter the dark compartment was 20 measured. A cutoff point of 300 sec was used for retention latency. Animals that failed to step through within 300 see were removed from the apparatus and a 300 sec latency was recorded for them. The tested compounds include: AF150(S) (0.5, 1 and 5 mg/kg, p.o.), AF267B (0.5, 1 and 5 mg/kg, p.o.) , AF102B 1 mg/kg, p.o.). The retention latency of trihexyphenidyl rats treated with AF150(S)- 5 mg/kg (222 - 25.6), AF267B- 0.5mg/kg (181.1 -35.4), AF267B- 1mg/kg (290.1 ±8.5) 25 and AF102B- 1mg/kg (234.4 135.3) was significantly longer than that of trihexyphenidyl rats treated with double distilled water (DDW) (82.9 -19.55) (p<0.01-0.001). Furthermore, the retention latency of the trihexyphenidyl rats treated with AF267B- 1 mg/kg was significantly longer than that of trihexyphenidyl rats treated with AF267B- 0.5 mg/kg (p<0.01) or trihexyphenidyl rats treated with AF150(S)- 5 mg/kg (p<0.0 5 ). No difference was found in the retention latency between control groups 30 treated with various drugs or DDW. AF704 was also significantly effective in this test. Thus retention latency of trihexyphenidyl rats treated with DDW (116.25-36.36) was significantly shorter than that of control (DDW) rats treated with DDW (300±0) (p<0.001). However, the retention latency of trihexyphenidyl rats treated with AF704-0.1 mg/kg, po (214.70-36.63), 0.5 mg/kg, po (283.50-17.39) and 1 mg/kg, po (274.44-26.97) was significantly longer than that of trihexyphenidyl rats treated with 35 DDW (p<0.01-0.001). AF292 was the most potent compound among the tested agonists. AF292 was significantly effective at a dose of 0.1-0.05 mg/kg, po. When the lowest dose of AF292 0.03 mg/kg, po was tested after 24 and 72 hrs, only the 72 hrs delay showed a significant effect on PA in retention latency {225.9±36 vs. DDW - 93.1± 29.0; p<0.01 compared to trihexyphenidyl rats treated with DDW). No WO 03/092580 PCT/IL03/00357 76 effects were found in the initial latency. These results show AF292 to be a highly potent agonist (e.g. more potent than AF150(S) by two orders of magnitudes), despite the higher potency of AF150(S) in binding studies against pirenzepine (high affinity & low affinity). This effect of AF292 cannot be attributted only to a higher biovailability of AF292 vs AF150(S) in rats (49% vs. 31%). 5 EXAMPLE 51 Effects of AF267B. AF292 on cognitive function in aged rats. Old (22-24 months old) and young (three months old) Sprague-Dawley rats had been tested in the MWM. Old rats showed a significantly slower learning curve than young rats, F(3/267)=6.74, 10 p<0.0001, and F(3/267)=4.66, p<0.
003 , for escape latency and path length, respectively. No significant effect was found for any of the test compounds on learning; however, aged rats treated with AF267B-1 mg/kg showed a tendency for improvement. All young rats showed a spatial bias in the probe trial, in both parameters, relative to old rats, F(3/267)=34.91, p<0.0001, and F(3/267)=9.06, p<0.0001, for escape latency and path length, respectively. No significant effect was found for any of the test 15 compounds on memory; however, relative to old rats treated with DDW, aged rats treated with AF292 in both doses, showed a tendency for partial spatial bias in this test. Old rats showed significantly worse performance during reversal learning than young rats. AF267B-1mg/kg improved significantly the reversal learning of aged rats, F(4/88)=2.62, p<0.04, and F(4/88)=2.58, p<0.0 4 , for escape latency and path length, respectively. The beneficial effects of 20 AF267B on reversal learning of old rats could not be attributed to nonspecific, motor coordination effects, since AF267B had no significant effect on the swimming ability of these rats. AF292 did not reach significance in the reversal learning of aged rats, yet from the shape of the curves there is a tendency of improvement at both doses tested, 1 and 0.5 mg/kg, po. 25 EXAMPLE 52 CNS Safety Profile of AF292 (Table 1) AF292 was evaluated in rodents for possible effects on general behavior and other CNS related pharmacological effects. No significant physical or behavioral signs were observed in rats administered AF292 at 1, 10, 30, 60, or 100 mg/kg orally, as compared to the vehicle control group. No behavioral 30 or physical signs were observed 24 hours after administration. All rats were retained for 14 days, and throughout this retention period all rats appeared normal. In comparison, the compound AF267B begins to show some effects (salivation and lacrimation at about 40 mg/kg po) already in the first hour after admistration.
WO 03/092580 PCT/IL03/00357 77 EXAMPLE 53 Cardiovascular Safety of AF292 1. Astemizole (human ether-a-go-go related gene (HERG) Channel) Binding Assay. AF292 was inactive in this binding assay as it failed to inhibit [ 3 H]-Astemizole binding to the hERG-encoded 5 channel. 2. Isolated Guinea Pig Right Atrium (atrial fibrillation). AF292 had no significant effect on contractile force, but tended to slightly reduce the contractile rate beyond what is seen in the vehicle group. 3. Effects of AF292 (dose 5 mg/kg orally) on cardiac electrophysiological, cardio- haemodynamic 10 in instrumented, awake dogs. Healthy trained and chronically instrumented female Beagle dogs of varying age and ranging in body weight from 9.4 to 12 kg, were used for recording of the cardiovascular parameters: heart rate, diastolic and systolic blood pressure, pressure rate product, LV dp/dt max, LV dp/dt max/pd, LV dp/dt min, cardiac output, stroke volume, systemic vascular resistance and the ECG parameters (PQ-, QRS-, QT-, QTcBazett- (QTcB), QTcFridericia- (QTcF) and 15 QTcVan de Water- (QTcVdW) interval duration and QT-dispersion. During the last 18 hours prior to the experiments, the dogs had no access to food. Water was available ad libitumn. At the beginning of each experiment, control values of the various parameters were recorded for at least 30 min. Thereafter, 5 mg/kg of AF292 (n=4) or the corresponding volume of the solvent (n=4) was administered orally by gavage. The various haemodynamic parameters were recorded continuously for 4 hours thereafter. 20 AF292 orally administered at a dose of 5 mg/kg has no statistically significant and relevant effect on hemodynamic or ECG parameters: e.g. blood pressure, cardiac contractility (LV dp/dt max; LV dp/dt max/pd) and relaxation (LV dp/dt min), stroke volume, systemic vascular resistance, the duration of the PQ-, QRS-, QT-, QTcB-, QTcF- and QTcVdW-interval, QT-dispersion and on ECG-morphology. 25 EXAMPLE 54 Effects of AF292 on CGtochrome P450 Isoform Inhibition Cytochrome P450 activity can be an indicator for potential drug-drug interactions. AF292 was evaluated in a microtiter plate assay for P450 inhibition. AF292, at a concentration up to 10 riM, did not 30 induce significant inhibition of CYP1A2, CYP2C9, CYP2C19, CYP3A4 and CPY2D6 isoforms, five major human P450 enzymes responsible for drug metabolism and associated drug-drug interactions. EXAMPLE 55 In vitro metabolizm of AF292 35 The metabolic stability of AF292 was evaluated by monitoring its disappearance while incubated with rat, rog, monkey, and human hepatic microsomes. Testosterone, and propranolol were run as assay controls. The results are shown in Table 1.
WO 03/092580 PCT/IL03/00357 78 EXAMPLE 56 Human colon adenocarcinoma (Caco-2) cell permeability studies of AF292. This test is used to determine intestinal permeability of tested compounds. Caco-2 cells, when grown on semipermeable filters, spontaneously differentiate in culture to form confluent monolayers 5 which both structurally and functionally resemble the small intestinal epithelium. Because of this property they are useful as an in vitro model for the study of drug absorption and metabolism during absorption in the intestinal mucosa. Caco-2 monolayers were grown to confluence on collagen-coated, microporous, polycarbonate membranes in 12 well plates and permeability of the test material was determined. The average permeability coefficient (Papp) of AF292 was 17.4X10 6 cm/sec ranking it as 10 having a high absorption potential (bi-directional assay performed). EXAMPLE 57 Protein binding Protein binding studies were carried out in human plasma, ax-glycoprotein, human serum 15 albumin (HSA), and Dulbecco's Phosphate Buffered Saline (PBS). AF292 was added to a final concentration of 10 pM. The results showed 0% protein binding in PBS buffer dose conc. (pM) and in human a-glycoprotein (AGP), (see Table 1). EXAMPLE 58 20 Pharmacokinetic profile of AF292 in rats and dogs The results of the PK profile of AF292 in rats and Beagle dogs (overnight fasting; drug administered in water solution; gavage) are listed in Table 1. EXAMPLE 59 25 Toxicology profile of AF267B AF267B has been extensively tested in chronic toxicity studies for up to 13 weeks in the Wistar rat and the Beagle dog. In the dog, the no-adverse-effect-level (NOAEL) is considered to be in the range of 6-9 mg/kg/day, po. Effects seen in the dog (>9 mg/kg) and rat (> 40 mg/kg) are consistent with the profile of AF267B as a muscarinic agonist, without toxic cardiovascular effects (tested in 30 awake dogs). AF267B was evaluated extensively in beagle dogs for up to 13 weeks in oral toxicity studies and electrocardiograms (ECG) were routinely recorderd pre- and post-administration of the test agent. Heart rate, P wave duration & amplitude, P-Q, QRS, and QT intervals were measured and no changes in the ECG considered to be related to the adminstration of the test agent were observed.
WO 03/092580 PCT/L03/00357 79 EXAMPLE 60 Effects of AF267B on rat hippocampal neurons exposed to A3 fibrils as followed by: survival and apoptosis: GSK-3p3 activity: cytoplasmic and nuclear stabilization of 3-catenin; cyclin D1 expression This study was performed in rat hippocampal primary cell cultures using the methods described 5 by Garrido JL et al. (FASEB J 2002; 16:1982). AF267B does not affect the survival and morphology of hippocampal neurons at concentrations of 0.5-50pM. However AF267B (10 pM) protects >90% the hippocampal neurons from A13 1-40 (5 pM) that alone caused a 45 % decrease in survival and in morphology. These protective effects of AF267B are mediated by M1 muscarinic receptors since these are blocked by pirenzepine (10 10 nM), an Ml antagonist. AF267B (100 pM) decreased GSK-3 3 activity by 60% in cultures of rat hippocampal neurons. In such a preparation 10 gM A3 fibrils increased GSK-3p3 activity to 370% vs control (100%). Furthermore AF267B (10pM) antagonized the effects of Ap fibrils (10 gM) decreasing GSK-3 3 activity to the same of 150% vs. control. AF267B (10 gM) prevented A3l1-40 (5 plM)-induced apotosis to the control level. When cultured hippocampal neurons were exposed to 15 A3pl-40 (5 pM), soluble P3-catenin was degraded and this degradation was prevented by AF267B (100 gM). In fact, AF267B increased soluble P-catenin level above control in a concentration dependent manner [e.g. 1 ±M (100%) 10 pM (300%), 100 gM (350%)]. AP31-40 (5 pM) decreased nuclear P-catenin (by 60%), an effect blocked by AF267B (1 yiM (140% vs control). These protective effects of AF267B are mediated by M1 mAChR since these are blocked by pirenzepine (10 rinM). The 20 destabilizing effect of AP3l-40 (5 pM) in rat hippocampal neurons was shown by dendritic shrinkage detected by immunofluorescent stain. The position of the nucleus was shown by c-jun antibody. AF267B protected the neurons and the cells have healthy neurites when treated with this M1 agonist. The effect of AF267B is Ml mAChR mediated since it is blocked by pirenzepine (10 nM). Finally AF267B has a protective effect (50% increase vs. control) against A131P-40 (5 PM)- induced decrease 25 (40% vs. control) of cyclin DI, a target gene of the Wnt pathway. Again this effect of AF267B is blocked by by pirenzepine (10 nMI). The results shown here indicate that AF267B protects neuronal cells as evaluated by MTT reduction, immunofluorescence ofneurofilaments and apoptotic analysis. 30 As shown above, compounds used in embodiments of the present invention are low molecular compounds that are capable of crossing the blood brain barrier. Many of these compounds have additional beneficial effects including, inter alia, improvement of memory and learning in a variety of animal models that mimic various aspects of AD and other related disorders with an excellent safety margin. 35 Table 1 compares some of the results of tests on AF292 and AF267B.
WO 03/092580 80 PCT/IL03/00357 Table 1 EFFECT AF292 AF267B Trihexyphenidyl-rats 0.03, 0.05, 0.1, 0.3, 0.5 mg/kg, p.o. 0.5, 1, 5 positive effects -Passive avoidance positive effects; IMIED</ 0.03 mg/kg, p.o. MED 0.1 - 0.5mg/kg, p.o. LONG DURATION OF ACTION CNS (rat): IRWIN TEST, Mydriasis: 25 mg/kg, p.o. General Observation 1, 10, 30, 60, 100 mg/kg, p.o. Salivation: 40 mg/kg, p.o. No effect on: Lacrimation: 50 mg/kg, p.o. motor activity {open field; vertical & Hypothermia: 50 mg/kg, p.o. horizontal screen; rotarod; locomotor ataxia; body Gnawing; 50 mg/kg, p.o. posture & tone; tremors; twiches; paralysis; Convulsion: 50 mg/kg, p.o. catalepsy} Sedation: 100 mg/kg, p.o. reflexes {righting; corneal; pinnmal; extension; limb Chromodacryorrhea: 100 mg/kg, p.o. tone; flexor withdrawal; startle) excitation or sedation {convulsion clonic/tonic; IRWIN TEST: 1. 10. 100 ma/k. p.o. opisthotonus; vocalization; C-tail; Straub tail; 1 & 10 no effect on gross behavior circling; stereotypies; sedation; hypnosis (sleep} & physiological state. eye condition {palpebral ptosis; lacrimation; 100 mg/kg: abnormalities of carriage, chromodacryorrhea; enophtalamus; exophtalamus} apathy, decreased corneal reflex, diarrhea, skin condition {skin plasticity, piloerection, reduced grooming, increased urination, blanching (ear); hyperemia, cyanosis} salivation, lacrimation & various effects Isalivation; diarrhea; diuresis; chromodacryorrhea, reduced locomotor response to handling;abdominal constriction; rectal activity, passivity, decreased respiration, temp.; death} reduced startle response (onset: 0.5 hr; duration 3-6 hrs.).
[
3 PZ]; rat cortex; Ki, 1.64+/-0.13 3.74+/-0.59 4M [1OXO-M]; rat 0.57+/-0.15 1.62+/-0.34 cortex, Ki, pM a-APPs secretion 100% Equipotent with AF267B & 100% in cell cultures carbachol.(CCh), EC50=3gM- Even though less transfected with rat efficacious on PI Ml mAChR (% of max CCh) MTT assay against Equipotent with AF267B & CCh at 100pM; Even 100% AP3 25-35 & H 2 0,2 though less efficacious on PI in cell cultures transfected with rat M1 mAChR PI turnover PI, AA: M1 mAChR: 35%, >88% PI, AA: Ml mAChR: 66%, 100% vs. CCh (as 100%) PI., AA: M3 mAChR: Not active as agonist PI: M3 mAChR: 30% in cell cultures An M3 antagonist (pKb=660 nM) PI, rat MI mAChR: 75% transfected M2 mAChR - no effects as agonist with the human PI, AA: M5 mAChR - no effect as agonist mAChR subtype PI, rat Ml mAChR; 50% Metabolic Stability Rat Liver Microsomes: T1/2 =92 min Liver Microsomes: TI/2 = 88 min Dog Liver Microsomes: T1/2> 100min Dog Liver Microsomes: T1/2> 100 min Monkey Liver Microsomes: T1/2 =74 min Monkey Liver Microsomes: T1/2=27 min Human Liver Microsomes: T1/2> 100 min Human Liver Microsomes: T1/2 >100 rain (BETTER THAN AF267B} WO 03/092580 81 PCT/IL03/00357 TABLE 1 CONT'D Protein Binding PBS= 0% PBS = 0% Human 0-glycoprotein (AGP) =0% AGP =5.7% Human serum albumin (HSA)= 22% HSA= 32% Human Plasma= 35% Human Plasma= 25% Pharmacokinetic Rats, 10 mg/kg, p.o. Dogs, 5mg/kg, p.o. Rats, 5 mg/kg, p.o. Dogs., 1mg/kg, p.o. study^ 0.99 2.04 T1/2, hr 0.5 0.8 0.64 1.33 Tmax, hr 1.5 4.85 0.25 0.58-0.75 MRT, hr 1906 1193 1 1.96 Cmax, ng/ml 2146 4648 852, 1704** 257, 1085# AUC(oine, (ng*hr/ml) 773, 1546** 552, 2760# Bioavailability (%F) 49 70 30*** 53 Toxicokinetic study: 13w* F (lx): outlier,3901-4840; M (lx):1142-10932 F (lx): 985-3179; M (lx): 800-3691 AUC(0-inf) ng*h/ml F(13w):1866-12723; M(13w):1609-4170 F(13w): 1033-4164; M(13w): 1041-4471 F (lx): outlier, 8.7-18.7; M (lx): 5-27.3 F (lx): 4.7-5.3; M (lx): 3.8-5 MRT (area) [h] F(13w):5.1-33.7; M(13w):5-7.8 F(13w):3.6-5.2; M(13w): 3.0-4.2 Mean T1/2 (M)= 6.3+/-5.9; Mean T1/2 (F)= 10.6+/-7.6 Mean T1/2 (M)= 2.1+/-0.6 Elimination half life Mean T1/2 (F)= 3.3+/-1.5 th1 *Dogs: AF267B {1.5, 3, 6 mg/kg, p.o.; single= lx & for 13 weeks= 13 w}; (AF292 obtained from AF267B, in vivo); treatment by gavage 1/2 h after feeding; solid drug in capsule. Plasma levels of the drug are analyzed at various time points by LC-MS; ** extrapolated for 10mg/kg; *** calculated for 2mg/kg, po; # (extrapolated for 5mg/kg). Oral and intravenous area under the concentration vs. time curve (AUC) were compared to determine the % biovailability (%/oF) by the following 5 formula: Dose (IV)xAUC (oral) / Dose (oral)xAUC(IV). A % F of over 30% generally suggests good bioavailability. Cmax levels are equally important to determine if sufficient plasma levels are attained to produce the desired pharmacological effect and a value > 1 iM is usually sufficient. AUMC is the first statistical moment of the AUC and is used to calculate the mean residence time (MRT=AUMC/AUC) which is the average time the compound is in the animal. The Cmax represents the maximum concentration observed, the Tmax is the time to reach that maximum concentration and the T1/2 is the calculated 10 half-life of the compound in plasma (ln2xMRT). Clearance is the volume of fluid (containing compound) from which compound is removed completely per unit time. ^Rat and dogs were dosed intravenously (iv) and by oral gavage. Plasma levels of the drug are analyzed at various time points by LC-MS-MS. @Irwin, S. PSYCHOPHARM 13:222-257,1968. The results shown in Table 1 indicate that AF267B and AF292 have the same affinity, but 15 differing efficacy for mAChR subtypes. It will be appreciated by persons skilled in the art that the present invention is not limited by what has been particularly shown and described hereinabove. Rather the scope of the present invention includes both combinations and subcombinations of the features described hereinabove as well as modifications and variations thereof which would occur to a person of skill in the art upon reading the 20 foregoing description and which are not in the prior art.

Claims (98)

  1. 54-130, unchanged, claims 131-173 new] We claim: 1. A compound of the formula (I): S/a b AC e (I 5 wherein: C denotes a spiro carbon atom shared by ring A and the ring containing a, b, d and e; A is selected from the group consisting of: RN C RN C -R§N C C C N N and 10 wherein R is selected from H, CI-C 8 straight- or branched-chain alkyl, or -CH 2 -P(=O)(OH) 2 ; a is -0- or -S-; b is -CR'R or -C(R )=; d is selected from the group consisting of =N-, -C(=0)-, -C(=S)- and =N(R3)=0; e is selected from the group consisting of -CH 2 -, -CHR 4 -, -Ni-, -NR 5 -, -N(SO 2 R 6 )- and 15 -N(C(=O)R 6 )-; R, R', R, R, R 4 are each independently selected from H, C1-6 alkoxy, C 2 . 6 hydroxyalkyl, C 2 -6 alkenyl, C 2 - 6 alkynyl, and C1-6 alkyl optionally substituted by one to three phenyls; R 5 is independently selected from H, C 1 _ alkyl optionally substituted by one, two or three phenyls, C 1 .6 alkoxy, C 2 -6 hydroxyalkyl, C 2 - 6 alkenyl, C 2 -6 alkynyl, optionally substituted phenyl, heteroaryl, and 20 C 1 . 6 alkyl optionally substituted by one to three phenyls; and R 6 is selected from C 1 -6 alkyl, C 1 - 6 alkoxy, C 1 -6 alkylthio, C 2 -6 hydroxyalkyl, C 2 . 6 alkenyl, C2-6 alkynyl, and C 3 - 7 cycloalkyl, each optionally substituted by from 1-6 halogen atoms, hydroxy-C 1 . 6 -alkyl, aryl substituted with a halogen, nitro, amino, hydroxyl, or CF 3 group, C 1 _6 alkyl substituted by one to three aryl groups, and Cm alkyl-X, wherein m=0 to 6 and X is selected from the 25 group consisting of indole, C 1 - 6 alkyl indole, isoindolyl, 3-pyridinyl, 3-piperidinyl, benzimidazolyl, thienyl, isothiazolyl, imidazolyl, pyrazinyl, benzofuranyl, quinolyl, isoquinolyl, benzothienyl, isobenzofuryl, pyrazolyl, indolyl, isoindolyl, benzimidazolyl, purinyl, carbazolyl, oxazolyl, thiazolyl, isothiazolyl, 1,2,5-thiadiazolyl, isooxazolyl, pyrrolyl, pyrazolyl, quinazolinyl, pyridazinyl, pyrazinyl, cinnolinyl, phthalazinyl, quinoxalinyl, xanthinyl, hypoxanthinyl, and pteridinyl; 30 or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof, WO 2003/092580 103 PCT/IL2003/000357 RN C with the proviso that when A is , R is -CH 3 , a is S, b is -CH(CH 2 CH 3 )- and d is -C(=O)-, then e is not -NHi- (AF267 or an enantiomer thereof), and with the further proviso that when A is RN C , R is -CH 3 , a is S, b is -C(CH 3 )= and d is =N-, then e is not -CH 2 - (AF150(S)). 5 2. A compound according to claim 1 wherein Rs 5 is heteroaryl selected from the group consisting of indole, pyrrolidinyl, piperidinyl, piperazinyl, furyl, pyridyl, pyrimidyl, thienyl, isothiazolyl, imidazolyl, pyrazinyl, benzofuranyl, quinolyl, isoquinolyl, benzothienyl, isobenzofuryl, pyrazolyl, indolyl, isoindolyl, benzimidazolyl, purinyl, carbazolyl, oxazolyl, thiazolyl, isothiazolyl, 1,2,5-thiadiazolyl, isooxazolyl, pyrrolyl, pyrazolyl, quinazolinyl, pyridazinyl, pyrazinyl, cinnolinyl, phthalazinyl, 10 quinoxalinyl, xanthinyl, hypoxanthinyl, pteridinyl, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and pyrazolopyrimidinyl. 3. A compound according to claim 1 which is a dimer of a compound of formula 1 wherein e is -NR 5 - and the two formula 1 moieties share a common group R 5 which is selected from the group 15 consisting of -(CH2),,- and -(CH 2 0),-, wherein n is I to 6, or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof. 4. A compound according to claim 1 selected from the group consisting of: N-[(2,8-Dimethyl 1-oxa- 8-aza-spiro[4.5]dec-3-ylidene)-amine]-N-oxide; N-[(2-Ethyl- 8-methyl- 1-oxa- 8-aza 20 spiro[4.5]dec- 3-ylidene)- amine]- N-oxide; N-[(2-Methyl- 8-phenyl- 1-oxa- 8-aza- spiro[4.5]dec 3-ylidene)- amine]- N-oxide; Thia-4,8-diaza- spiro[4.5]decan- 3-one; 4-(2,4-Dimethoxybenzyl) 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF286); 8-Methyl- 4-pyrrolidin- 1-ylmethyl 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF287); 2-(1-Hydroxy-ethyl)- 8-methyl- 1-thia- 4,8-diaza spiro[4.5]decan- 3-one (AF298); (S)-2-Ethyl- 8-methyl- 8-oxy- 1-thia- 4,8-diaza- spiro[4.5]decan 25 3-one (AF299); 4-(2,4-Dimethoxy-benzyl)- 2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF288); (S)-2-Ethyl- 8-methyl- 1-oxo- 1. 4 -thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF300); 2-Ethyl 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decane- 3-thione (AF510); (S)-2-Ethyl 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF700); 2-Ethyl 4-[2-(1H-indol-3-yl)-ethyl]- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF703); 2-Ethyl 30 4-(3-IH-indol-3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF704); (S)-Ethyl 4-(3-IH-indol- 3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF704B); (R)-Ethyl- 4-(3-lH-indol- 3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF704A); and 2-Methyl- 8-methyl-d 3 -1-thia- 3,8-diaza- spiro[4.5]dec- 2-ene (AF402), or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, metabolite, or a 35 pharmaceutically acceptable salt thereof. WO 2003/092580 PCT/IL2003/000357 104 RN C 5. A compound according to Claim 1 wherein wherein A is , R is H, a is -S-; b is -CH(CH 2 CH 3 )-; d is -(C=O)-; and e is -NH-, i.e. 2-Ethyl-I -thia-4,8-diaza-spiro[4.5]decan-3-one (AF504). or an enantiomer, diastereomer, geometrical isomer, racemate, tautomer, dimer, metabolite or 5 pharmaceutically acceptable salt thereof 6. A compound according to claim 5 which is (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF292) or a pharmaceutically acceptable salt thereof. 10 7. A compound according to claim 6 which is the HCI salt of AF292. 8. The compound (S)-2-Ethyl-1-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292) according to claim 1, or a metabolite or pharmaceutically acceptable salt thereof, whenever located in a human or animal body. 15 9. A compound according to claim 5 which is (R)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF291) or a pharmaceutically acceptable salt thereof. RN C 10. A compound according to Claim I wherein A is , R is -CH 3 , a is -0-, d is 20 =N(R 3 )=0, or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof. 11. A compound according to claim 10 wherein b is -CH(CH 3 )- and R 3 is -CH 3 , i.e. N-[(2,8-Dimethyl- I -oxa-8-aza-spiro[4.5]dec-3-ylidene)-methyl-amine]-N-oxide (AF600). 25 12. A compound according to claim 10 wherein b is -CH(CH 3 )-, R 3 is benzyl, i.e. N-[(2,8-Dimethyl-I-oxa-8-aza-spiro[4.5]dec-3-ylidene)-benzyl-amine]-N-oxide (AF604) 13. A compound according to claim 10 wherein b is -CH(CH 3 )- and R 3 is isopropyl, i.e. 30 N-[(2,8-Dimethyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene)- isopropyl-amine]- N-oxide (AF605) 14. A compound according to claim 10 wherein b is -CH(CH 2 CH 3 )- and R 3 is -CH 3 , i.e. N-[(2-Ethyl 8-methyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene)- methyl- amine]- N-oxide (AF601). WO 2003/092580 105 PCT/IL2003/000357 15. A compound according to claim 10 wherein b is -CH(CH 3 )- and R 3 is phenyl, i.e. N-[(2-Methyl 8-phenyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene)- methyl-amine]- N-oxide (AF602). 16. A compound according to Claim I wherein A is CO , R is -CH 3 , a is -0-, d is 5 =N(R 3 )=0, or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof. 17. A compound according to claim 16 wherein b is -CH(CH 3 )- and R 3 is methyl, i.e. Dihydro 5'-methylspiro [1-azabicyclo[2.2.2]octane- 3,5'-(4'H)- 3'-ylidene- methylamine]-N-oxide (AF603). 10 RNC 18. A compound according to Claim 1 wherein wherein A is \ , R is methyl, a is -S-, b is -CH(CH 2 CH 3 )-; d is -C(=O)-; e is -NR 5 - wherein R 5 is selected from -(CH 2 ) 3 -indolyl and -C(=O)-(CH 2 ) 3 -indolyl, i.e. 2-Ethyl- 4-[2-(1H-indol-3-yl)-ethyl]- 8-methyl- 1-thia- 4,8-diaza spiro[4.5]decan-3-one (AF703) or 2-Ethyl- 4-(3-1H-indol-3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza 15 spiro[4.5]decan-3-one (AF704), or an enantiomer, diastereomer, geometrical isomer, racemate, tautomer, dimer, metabolite or pharmaceutically acceptable salt thereof. 19. A compound according to claim 18 which is (S)-Ethyl-4-(3-1H-indol- 3-yl-propionyl) 8-methyl- 1-thia-4,8-diaza-spiro[4.5]decan-3-one (AF704B). 20 RN C 20. A compound according to Claim 4 wherein wherein A is , R is methyl, a is -S-, b is -CH(CH 2 CH 3 )-, d is -C(=O)-, e is -N(SO 2 R 6 )- and R 6 is 4-fluoro-benzene, and the 2-carbon has the S-configuration, i.e. (S)-2-Ethyl- 4-(4-fluoro- benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza spiro[4.5]decan- 3-one (AF700). 25 21. A pharmaceutical composition comprising at least one compound according to any one of claims 1 to 20, or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient therefor. 30 22. A pharmaceutical composition according to claim 21, further comprising at least one additional compound chosen from the group consisting of (S)-2-ethyl- 8-methyl- 1-thia- 4,8-diaza spiro[4.5]decan- 3-one (AF267B) and 2,8-Dimethyl- 1-thia- 3,8-diaza- spiro[4.5] dec- 2-ene (AF1 50(S)), or a pharmaceutically acceptable salt thereof. WO 2003/092580 .PCT/IL2003/000357 106 23. A pharmaceutical composition according to claim 21, wherein said at least one compound is (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF292) or a prodrug of AF292 or a pharmaceutically acceptable salt thereof. 5 24. A pharmaceutical composition according to claim 23, wherein said prodrug is (S)-2-ethyl 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF267B) or a pharmaceutically acceptable salt thereof 10 25. A pharmaceutical composition according to claim 21 comprising an MI muscarinic receptor antagonistic amount of at least one compound selected from the group consisting of: RN C compounds of formula (I) wherein A is , R is -CH 3 , a is -0-, d is N(R 3 )=O; N compounds of formula (I) wherein A is C , R is -CH 3 , a is -0-, d is =N(R 3 )=0; 2-Ethyl 4-[2-(1H-indol-3-yl)-ethyl]- 8-methyl- I-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF703); and 2-Ethyl 15 4-[2-(1H-indol-3-yl)-ethyl]- 8-methyl- I-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF703) or 2-Ethyl 4-(3-lH-indol- 3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF704). 26. A pharmaceutical composition according to claim 23, wherein said at least one compound is (S)-2-Ethyl- I-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF292) or a pharmaceutically acceptable salt 20 thereof in an MI muscarinic receptor agonistic and M3 muscarinic receptor antagonistic amount. 27. A pharmaceutical composition according to claim 23, wherein said at least one compound is (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF292) or a pharmaceutically acceptable salt thereof, further comprising at least one additional compound selected from the group consisting of 25 compounds of formula (I) according to claim 1, AF267B and AFl50(S). 28. A pharmaceutical composition according to claim 23, further comprising at least one additional compound which is (S)-2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF267B) or a pharmaceutically acceptable salt thereof. 30 29. A pharmaceutical composition according to claim 23, further comprising at least one additional compound which is 2,8-Dimethyl- 1-thia- 3,8-diaza- spiro[4.5] dec- 2-ene (AF150(S)) or a pharmaceutically acceptable salt thereof. 35 30. A pharmaceutical composition according to claim 23 comprising an MI muscarinic receptor agonistic and M3 muscarinic receptor antagonistic amount of (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] WO 2003/092580 PCT/IL2003/000357 107 decan- 3-one (AF292) or a pharmaceutically acceptable salt thereof, or a combination of AF292 or a pharmaceutically acceptable salt thereof and (S)-2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan 3-one (AF267B) or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. 5 31. A pharmaceutical composition according to claim 23 comprising a schizophrenia treating amount of (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan- 3-one (AF292) or a pharmaceutically acceptable salt thereof, or a combination of AF292 or a pharmaceutically acceptable salt thereof and (S)-2-ethyl 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF267B) or a pharmaceutically acceptable salt 10 thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. 32. A pharmaceutical composition according to claim 23 comprising a schizophrenia symptom ameliorating amount of (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF292) or a pharmaceutically acceptable salt thereof, or a combination of AF292 or a pharmaceutically acceptable 15 salt thereof and (S)-2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF267B) or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. 33. A pharmaceutical composition according to claim 21, wherein said at least one compound is 20 selected from the group consisting of: N-[(2,8-Dimethyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene) amine]-N-oxide; N-[(2-Ethyl- 8-methyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene)- amine]-N-oxide; N-[(2-Methyl- 8-phenyl- 1-oxa- 8-aza- spiro[4.5]dec-3-ylidene)- amine]-N-oxide; thia-4,8-diaza spiro[4.5]decan- 3-one; 4-(2,4-Dimethoxybenzyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF286); 8-Methyl- 4-pyrrolidin- 1-ylmethyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF287); 25 2-(1-Hydroxy-ethyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF298); (S)-2-Ethyl 8-methyl- 8-oxy- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF299); 4-(2,4-Dimethoxy-benzyl) 2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF288); (S)-2-Ethyl- 8-methyl- 1-oxo 1 4 -thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF300); 2-Ethyl- 8-methyl- 1-thia- 4,8-diaza spiro[4.5]decane- 3-thione (AF510); (S)-2-Ethyl- 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia 30 4,8-diaza- spiro[4.5]decan- 3-one (AF700); 2-Ethyl- 4-[2-(IH-indol-3-yl)-ethyl]- 8-methyl- 1-thia 4,8-diaza- spiro[4.5]decan- 3-one (AF703); 2-Ethyl- 4-(3-1H-indol-3-yl-propionyl)- 8-methyl- 1-thia 4,8-diaza- spiro[4.5]decan- 3-one (AF704); (S)-Ethyl- 4-(3-lH-indol- 3-yl-propionyl)- 8-methyl 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF704B); (R)-Ethyl- 4-(3-1H-indol- 3-yl-propionyl) 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan- 3-one (AF704A); 2-Methyl- 8-methyl-d 3 - 1-thia 35 3,8-diaza- spiro[4.5]dec- 2-ene (AF402), 2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF504), (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF292), (R)-2-Ethyl- 1-thia- 4,8-diaza spiro[4.5]decan- 3-one (AF291), N-[(2,8-Dimethyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene)- methyl amine]- N-oxide (AF600), N-[(2,8-Dimethyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene)- benzyl- amine] N-oxide (AF604), N-[(2,8-Dimethyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene)- isopropyl- WO 2003/092580 PCT/IL2003/000357 108 amine]-N-oxide (AF605), N-[(2-Ethyl- 8-methyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene) methyl-amine]- N-oxide (AF601), N-[(2-Methyl- 8-phenyl- 1-oxa- 8-aza- spiro[4.5]dec- 3-ylidene) methyl- amine]- N-oxide (AF602), dihydro- 5'-methylspiro [1-azabicyclo[2.2.2]octane- 3,5'-(4'H) 3'-ylidene- methylamine]- N-oxide (AF603), 2-Ethyl- 4-[2-(1H-indol- 3-yl)-ethyl]- 8-methyl- 1-thia 5 4,8-diaza- spiro[4.5]decan- 3-one (AF703), 2-Ethyl- 4-(3-1H-indol- 3-yl-propionyl)- 8-methyl- 1-thia 4,8-diaza- spiro[4.5]decan- 3-one (AF704), (S)-Ethyl- 4-(3-1H-indol- 3-yl-propionyl)- 8-methyl- 1-thia 4,8-diaza- spiro[4.5]decan- 3-one (AF704B), and (S)-2-Ethyl- 4-(4-fluoro- benzenesulfonyl)- 8-methyl 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF700), or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof. 10 34. A pharmaceutical composition according to claim 33, wherein said compound is AF700. 35. A pharmaceutical composition according to claim 33, wherein said compound is AF704B. 15 36. A pharmaceutical composition according to claim 33, further comprising at least one additional compound chosen from the group consisting of 2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan 3-one (AF267) and 2,8-Dimethyl- 1-thia- 3,8-diaza- spiro[4.5] dec- 2-ene (AF150(S)), or a pharmaceutically acceptable salt thereof. 20 37. A pharmaceutical formulation comprising 2,8-Dimethyl- 1-thia- 3,8-diaza- spiro[4.5] dec- 2-ene (AF150(S)) in paraffin oil. 38. A pharmaceutical composition comprising (a) an efficacious amount of a compound selected from the group consisting of (S)-2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF267B), 25 (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5]decan-3-one (AF292), and (S)-Ethyl- 4-(3-1H-indol 3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF704B), pharmaceutically acceptable salts thereof or mixtures of such compounds or salts, and (b) an efficacious amount of a compound selected from the group consisting of an immunotherapeutic compound against beta-amyloids and compounds that bind to amyloids, and a pharmaceutically acceptable carrier, diluent 30 or exicipient therefor. 39. A pharmaceutical composition according to any one of claims 21 to 38, wherein said pharmaceutical composition is in unit dosage form. 35 40. A pharmaceutical composition according to any one of claims 21 to 39, wherein said pharmaceutical composition is a powder, tablet, pill, capsule, cachet, suppository, a plurality of dispersible granules, a solution, a suspension, or an emulsion. WO 2003/092580 PCT/IL2003/000357 109 41. A pharmaceutical composition according to claim 39, wherein said pharmaceutical composition comprising between about 0.5 and about 100 mg of a compound of formula (I) per unit dosage form. 42. A pharmaceutical composition according to claim 41, wherein said pharmaceutical composition 5 comprising between about 5.0 and about 100 mg of a compound of formula (I) per unit dosage form. 43. A pharmaceutical composition according to claim 42, wherein said pharmaceutical composition comprising between about 10 and about 50 mg of a compound of formula (I) per unit dosage form. 10 44. A pharmaceutical composition according to any one of claims 21 to 40, wherein said pharmaceutical composition is orally, transdermally, bucally, intranasally, intraperitoneally, intravenously, rectally, parenterally or subcutaneously administrable. 45. A pharmaceutical composition according to claim 21 comprising at least one compound or a 15 mixture of compounds selected from the group consisting of: compounds of formula (I) wherein A is RN C ,R is -CH 3 , a is -0-, d is =N(R 3 )=O; compounds of formula (I) wherein A is N C g, R is -CH 3 , a is -0-, d is =N(R 3 )=0; 2-Ethyl- 4-[2-(1H-indol-3-yl)-ethyl]- 8-methyl 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF703); 2-Ethyl- 4-(3-1H-indol-3-yl-propionyl)- 8-methyl 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF704); AF267(B); AF150(S); or racemates, enantiomers, 20 geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in an amount efficacious to elevate the level of secreted form of the non-amyloidogenic amyloid precursor protein (ca-APPs), decrease the level of AP3 peptide in the brain of a mammal having an elevated level of AP3 in the brain, inhibit the release or synthesis of ApoE, decrease levels of ApoE, decrease tau hyperphosphorylation, decrease paired helical formation, activate the Wnt signaling pathway, increase 25 beta-catenin, inhibit GSK3-mediated effects or enhance the activity of neurotrophins. 46. A pharmaceutical composition comprising at least one compound selected from the group consisting of compounds of formula (I) according to any one of Claims 1-8, AF267B and AFI50(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable 30 salts thereof and at least one additional pharmacologically active compound selected from the group constisting of: cholinesterase inhibitors, nicotinic agonists, cholinergic precursors and cholinergic enhancers, nootropics, peripheral antimuscarinc drugs, M2 muscarinic antagonists, M4 antagonists, benzodiapine inverse agonists, antidepressants, tricyclic antidepressents or antimuscarinic drugs used in treatment of Parkinson's disease (PD) or depression, antipsychotic and antischizophrenic agents, 35 glutamate antagonists and modulators, NMDA antagonists, AMPA agonists, acetyl-L-carnitine, WO 2003/092580 PCT/IL2003/000357 110 MAO-B inhibitors, peptides and growth factors, cholesterol-lowering agents, antioxidants, GSK-313 inhibitors, Wnt-ligands, 3- or y-secretase inhibitors, beta-amyloid degrading agents, beta-amyloid anti-aggregation agents, chelating agents, immunotherapeutic compounds against beta-amyloids, compounds that bind to amyloids, cyclooxygenase (COX)-2 inhibitors, non-steroidal 5 antiinflammatory drugs, estrogenic agents, estrogenic receptor modulators, steroidal neuroprotectants, and spin trapping pharmaceuticals. 47. A pharmaceutical composition according to claim 46, wherein said at least one compound is AF292 or a prodrug of AF292 or a pharmaceutically acceptable salt thereof. 10 48. A pharmaceutical composition comprising an Ml muscarinic receptor stimulating and oa secretase activating efficacious amount of a compound of formula (I) according to claim 1 which is (S)-2-Ethyl 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF700), or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, 15 and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. 49. A pharmaceutical composition comprising an M1 muscarinic receptor agonistic and P3 secretase antagonistic efficacious amount of a compound of formula (I) according to claim 1 which is (S)-2-Ethyl- 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one 20 (AF700), or an enantiomer, diastereomer, geometrical isomer, racemate, tautomer, dimer, metabolite or pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. 50. A pharmaceutical composition comprising an MI muscarinic receptor agonistic and y secretase 25 antagonistic efficacious amount of a compound of formula (I) according to claim 1 which is (S)-2-Ethyl- 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF700), or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. 30 51. A pharmaceutical composition comprising an Ml muscarinic receptor agonistic and M3 muscarinic receptor antagonistic efficacious amount of the compound (S)-2-Ethyl-1-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292), or a metabolite or pharmaceutically acceptable salt thereof, and at least one pharmaecutically acceptable carrier, diluent or excipient 35 therefor. 52. A pharmaceutical composition comprising an Ml muscarinic receptor agonistic and M3 muscarinic receptor antagonistic amount of the compound (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF292) or a pharmaceutically acceptable salt thereof, or a combination of the compounds WO 2003/092580 PCT/IL2003/000357 111 (S)-2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decan- 3-one (AF267B) or a pharmaceutically acceptable salt thereof and (AF292) or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. 5 53. A pharmaceutical composition comprising an MI muscarinic receptor agonistic amount of a combination of (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF292) or a pharmaceutically acceptable salt thereof, or a combination of a first compound which is (AF292) or a pharmaceutically acceptable salt thereof and a second compound selected from the group consisting of compounds according to claim 1, AF267B and AF150(S), including racemates, enantiomers, diastereomers, 10 geometric isomers, tautomers and pharmaceutically acceptable salts thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient therefor. 54. A pharmaceutical composition comprising an Ml muscarinic receptor stimulating, oxidation-retarding and neuroprotectant activity efficacious amount of the compound 2-Ethyl 15 4-(3-1H-indol- 3-yl-propionyl)- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one in racemic form (AF704) or as the S-enantiomer thereof (AF704B), and a pharmaceutically acceptable carrier, diluent, or excipient therefor.
  2. 55. A pharmaceutical composition comprising for inhibiting the release or synthesis of beta-amyloid 20 peptide (A3) in a mammal comprising an amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to inhibit the cellular release or synthesis of A3. 25 56. A pharmaceutical composition for elevating the level of secreted form of the non-amyloidogenic amyloid precursor protein (a-APPs) in a mammal comprising an amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to elevate the level of the secreted form of the 30 non-amyloidogenic amyloid precursor protein (c-APPs).
  3. 57. A pharmaceutical composition for decreasing the level of A3 peptide in the brain of a mammal having an elevated level of A3 in the brain, comprising an amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to claim 1, 35 AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to decrease the level of A3 in the brain of said mammal. WO 2003/092580 112 PCT/IL2003/000357
  4. 58. A pharmaceutical composition according to claim 57 wherein said elevated level of AP in the brain is a result of hypercholesterolemia.
  5. 59. A pharmaceutical composition according to claim 57 wherein said elevated level of AP3 in the 5 brain is a result of cholinergic hypofunction.
  6. 60. A pharmaceutical composition for inhibiting the synthesis or release of apolipoprotein (ApoE) in a mammal comprising an amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), or racemates, 10 enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to inhibit the release or synthesis of ApoE in said mammal.
  7. 61. A pharmaceutical composition according to claim 60 wherein said ApoE is ApoE4. 15 62. A pharmaceutical composition for decreasing levels of apolipoprotein (ApoE) in a mammal comprising an amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), or racemates, geometrical isomers, enantiomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to decrease the levels of ApoE in said mammal. 20
  8. 63. A pharmaceutical composition according to claim 62 wherein said ApoE is ApoE4.
  9. 64. A pharmaceutical composition for decreasing tau hyperphosphorylation in a mammal, comprising a compound or a mixture of compounds selected from the group consisting of compounds of formula 25 (I) according to claim 1, AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to inhibit tau hyperphosphorylation.
  10. 65. A pharmaceutical composition according to claim 64 wherein said tau hyperphosphorylation is 30 AP3-induced tau hyperphosphorylation.
  11. 66. A pharmaceutical composition for decreasing paired helical formation in a mammal comprising a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, 35 diasteromers, tautomers and pharmaceutically acceptable salts thereof effective to inhibit tau hyperphosphorylation.
  12. 67. A pharmaceutical composition for activating the Wnt signaling pathway in a mammal, comprising a compound or a mixture of compounds selected from the group consisting of compounds WO 2003/092580 PCT/IL2003/000357 113 of formula (I) according to claim 1, AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers, geometrical isomers and pharmaceutically acceptable salts thereof effective to inhibit Wnt abnormalities. 5 68. A pharmaceutical composition for inhibiting GSK3 P-mediated effects in a mammal comprising a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers, geometrical isomers and pharmaceutically acceptable salts thereof effective to inhibit GSK3 -mediated effects. 10
  13. 69. A pharmaceutical composition according to claim 68 wherein said GSK313-mediated effects are selected from the group consisting of tau hyperphosphorylation, apoptosis, P-catenin degradation, and decrease in Wnt target genes. 15 70. A pharmaceutical composition for enhancing the activity of endogenous growth factors, i.e. neutrophins, comprising an amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof which alone is effective as a neurotrophic agent and which acts synergistically with said 20 neurotrophins.
  14. 71. A pharmaceutical composition for inhibiting the release or synthesis of beta-amyloid peptide (AP3), for elevating the level of secreted form of the non-amyloidogenic amyloid precursor protein (x-APPs), for decreasing the level of AP3 peptide in the brain of a mammal having an elevated level of 25 AP3 in the brain, for inhibiting the release or synthesis of ApoE, for decreasing levels of ApoE, for decreasing tau hyperphosphorylation, for decreasing paired helical formation, for activating the Wnt signaling pathway, for increasing beta-catenin, for inhibiting GSK313-mediated effects or for for enhancing the activity of neurotrophins, comprising a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), or 30 racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient.
  15. 72. A pharmaceutical composition for treating or reducing cerebral amyloid angiopathy comprising (a) an efficacious amount of a compound selected from the group consisting of AF267B, AF292, and 35 AF704B, pharmaceutically acceptable salts thereof or mixtures of such compounds or salts, and (b) an efficacious amount of a compound selected from the group consisting of an immunotherapeutic compound against beta-amyloids and compounds that bind to amyloids, and a pharmaceutically acceptable carrier, diluent or excipient. ... .. I , nrl"rIcin\ WO 2003/092580 PCT/IL2003/000357 114
  16. 73. A pharmaceutical composition for treating in a mammal diseases associated with impaired cholinergic function or diseases where there is an imbalance in cholinergic function, or diseases with impared activity of acetylcholine receptors from the group consisting of: senile dementia of Alzheimer's type; Alzheimer's disease (AD); Lewy body dementia; mixed Alzheimer's and Parkinson's disease; 5 multiifract dementia (MID); fronto-temporal dementia; vascular dementia; stroke/ischemia, MID combined with stroke/ischemia/head injury; combined MID and AD; human head injury; age-associated memory impairments; mild cognitive impairment (MCI); MCI conducive to AD; cognitive dysfunction (including forgetfulness, acute confusion disorders, attention-deficit disorders, focus and concentration disorders); hallucinatory-paranoid states; emotional and attention disorders; 10 sleep disorders; post-operative delirium; adverse effects of tricyclic antidepressants; adverse effects of certain drugs used in the treatment of schizophrenia and Parkinson's disease; xerostomia, anomia, memory loss and/or confusion; psychosis; schizophrenia, schizophrenia comorbit with AD, late onset schizophrenia, paraphrenia, schizophreniform disorders; anxiety; bipolar disorders; mania; mood stabilization; cognitive impairments after removal of certain gliomas; tardive dyskinesia; oxidative 15 stress during oxygen therapy; aphasia; postencephalitic amnesic syndrome; AIDS dementia; memory impairments in autoimmune diseases including lupus, multiple sclerosis, Sjogren's syndrome, chronic fatigue syndrome, and fibromyalgia; memory impairments in atypical depression or schizophrenia; pain, rheumatism, arthritis and terminal illness;, xerophtalmia, vaginal dryness, skin dryness; immune dysfunctions; neurocrine disorders and dysregulation of food intake, including bulimia and anorexia; 20 obesity; congenital ornithine transcarbamylase deficiency; ollivopontocerebral atrophy; alcohol withdrawal symptoms; substance abuse including withdrawal symptoms and substitution therapy; Huntington's chorea; progressive supranuclear palsy; Pick's disease; Friedrick's ataxia; Gilles de la Tourette disease; Down's syndrome; glaucoma; presbyopia; autonomic disorders including dysfunction of gastrointestinal motility and function such as inflammatory bowel disease, irritable bowel syndrome, 25 diarrhea, constipation, gastric acid secretion and ulcers; urinary urge incontinence, asthma, COPD; comprising an efficacious amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to any one of claims 1-18, AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof and a pharmaceutically acceptable carrier, diluent or excipient. 30
  17. 74. A pharmaceutical composition for preventing or treating central or peripheral nervous system disease states due to dysfunction in one or more of the following: brain, nervous system, cardiovascular system, immune system, neurocrine system, gastrointestinal system, or endocrine and exocrine glands, eye, cornea, lungs, prostate, or other organs where the cholinergic function is mediated by muscarinic 35 receptor subtypes, wherein said dysfunction involves: brain amyloid-mediated disorders; glycogen synthase kinase (GSK3p)-mediated disorders; tau protein hyperphosphorylation-mediated damages, dysfunctions or diseases; CNS and PNS hypercholesterolemia- and/or hyperlipidemia-mediated damages, dysfunctions or diseases; Wnt-mediated signaling abnormalities; impairment of neuroplasticity; hyperglycemia; diabetes; endogenous growth factors-mediated diseases, or WO 2003/092580 PCT/IL2003/000357 115 combination of additional risk factors; or disease states that involve apolipoprotein E; or disturbances in which a cholinergic dysfunction has been implicated, including: senile dementia of Alzheimer's type, Alzheimer's disease (AD), delay of onset of AD symptoms in a patient at risk for developing AD, Lewy body dementia, cerebral amyloid angiopathy (CAA), cerebral amyloidosis, fronto-temporal dementia, 5 vascular dementia, hyperlipidemia, hypercholesterolemia, multiifract dementia (MID), stroke ischemia, MID combined with stroke/ischemia/head injury, combined MID and Alzheimer's disease, human head injury, age-associated memory impairments, mild cognitive impairment (MCI), MCI conducive to AD, bipolar disorder, mania, schizophrenia, nonaffective sychozophrenia, paraphrenia, immune dysfunctions, neurocrine disorders and dysregulation of food intake, including bulimia and anorexia, 10 weight control, obesity, inflammation; comprising an efficacious amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to any one of claims 1-8, AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof and a pharmaceutically acceptable carrier, diluent or excipient. 15
  18. 75. A pharmaceutical composition according to claim 73 or 74, wherein said compound is AF292 or a prodrug of AF292 or a pharmaceutically acceptable salt thereof.
  19. 76. A pharmaceutical composition for treating a patient with AD, MCI, Lewi Body Dementia, 20 fronto-temporal dementia, vascular dementia, memory impairment in head injury, AIDS dementia in order to inhibit further deterioration in the condition of said patient comprising an efficacious amount of a compound or a mixture of compounds selected from the group consisting of compounds of formula (I) according to any one of claims 1-8, AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers, geometric isomers and pharmaceutically acceptable salts thereof, and a 25 pharmaceutically acceptable carrier, diluent or excipient.
  20. 77. A pharmaceutical composition for the treatment of schizophrenia, comprising AF267B, AF292, pharmaceutically acceptable salts thereof or a mixture of AF267B, AF292 or pharmaceutically salts thereof and at least one pharmaceutically acceptable carrier, diluent or excipient. 30
  21. 78. A pharmaceutical composition for amelioration of symptoms of schizophrenia, comprising a compound selected from AF267B, AF292, pharmaceutically acceptable salts thereof and mixtures of AF267B, AF292 or pharmaceutically acceptable salts thereof and. at least one pharmaceutically acceptable carrier, diluent or excipient. 35
  22. 79. Human or animal blood containing the compound (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF292), or a metabolite or pharmaceutically acceptable salt thereof. WO 2003/092580 PCT/IL2003/000357 116
  23. 80. Human or animal blood according to claim 79, wherein said blood is located in a human or animal body.
  24. 81. Human or animal blood according to claim 79, wherein said blood is not located in a human or 5 animal body.
  25. 82. Human or animal blood plasma containing the compound (S)-2-Ethyl-l-thia- 4,8-diaza-spiro[4.5] decan-3-one (AF292), or a metabolite or pharmaceutically acceptable salt thereof. 10 83. Human or animal blood plasma according to claim 82, wherein said blood plasma is located in a human or animal body.
  26. 84. Human or animal blood plasma according to claim 82, wherein said blood plasma is not located in a human or animal body. 15
  27. 85. A process for the preparation of 2-ethyl-8-methyl-1-thia-4,8-diaza- spiro[4.5]decan-3-one (AF267), comprising reacting 4-ethyl piperidone with 2-mercaptobutyric acid and ammonia.
  28. 86. A process according to claim 85, further comprising obtaining the enantiomers AF267A 20 (R-enantiomer) and AF267B (S-enantiomer) by chiral separation.
  29. 87. A process for the preparation of AF267B, comprising racemizing AF267A and isolating AF267B from the racemic mixture. 25 88. A process according to claim 87, wherein said isolating comprising separating the AF267B from the racemic mixture by chiral separation.
  30. 89. A process for the synthesis of AF267B comprising contacting (R)-2-mercaptobutyric acid with ammonium acetate and 1 -methyl-4-piperidone. 30
  31. 90. A process according to claim 89, wherein said (R)-2-mercaptobutyric is obtained by contacting (R)-2-benzoylthiobutyric acid with ammonium hydroxide.
  32. 91. A process according to claim 90, wherein said (R)-2-benzoylthiobutyric acid is obtained by 35 contacting (R)-2-bromobutyric acid with cesium thiobenzoate.
  33. 92. A process according to claim 90, wherein said (R)-2-bromobutyric acid is obtained by contacting 2-aminobutyric acid having the R configuration with sodium nitrite, potassium bromide and hydrobromic acid. WO 2003/092580 PCT/IL2003/000357 117
  34. 93. A process for the preparation of AF267B isotopically labelled with 13 C or "'C, comprising reacting AF287 with 13C- or 14C-labelled ethyl bromide, deprotecting the nitrogen atom at the 4-position of the AF287, and isolating " 3 C- or 1 4 C-labelled AF267B by chiral chromatography. 5
  35. 94. A process for the preparation of a mixture of (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF292) and (R)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF291) comprising reacting AF267 with a demethylating agent. 10 95. A crystalline form of (S)-2-ethyl-8-methyl-1-thia-4,8-diaza- spiro[4.5]decane -3-one (AF267B) characterized by the following data: P212121 (No 16) a=10.394 ((10) (at=90 0 ), b=20.133(2) (P=90 0 ), c=5.856 (4) (7=90o) A, T=1 10K.
  36. 96. A crystalline form according to claim 80, further characterized by the following data: Volume = 15 1224.2 (9) A 3 , Z = 4, Fw = 202.32, Calculated density, De = 1.092Mg/m 3 , Absorption coefficient, g = 0.232mm - .
  37. 97. A crystalline form of (S)-2-ethyl- 8-methyl- 1-thia- 4,8-diaza- spiro[4.5]decane- 3-one (AF267B) having the configuration shown in Structure I in Example 2. 20
  38. 98. A process for the preparation of 2,8-Dimethyl- 1-thia- 3,8-diaza- spiro[4.5]dec- 2-ene [AFl50(S)] comprising cyclizing 1-methyl- 4-N-thioacetylamino- 1,2,3,6-tetra- hydropyridine.
  39. 99. A process according to claim 98 wherein said cyclizing is conducted in the presence of phosphoric 25 acid.
  40. 100. A process according to claim 98 wherein said 1- methyl- 4-N-thioacetylamino 1,2,3,6-tetrahydropyridine is obtained by reduction of reduction of 1-methyl 4-N-thioacetylaminomethyl pyridinium with sodium borohydride. 30
  41. 101. A process according to claim 98 wherein said 1-methyl- 4-N-thioacetylaminomethyl pyridinium is obtained by reacting 4-(acetaminomethyl)- 1-methyl-pyridinium iodide with Lawesson's reagent.
  42. 102. A pharmaceutical formulation comprising 2,8-Dimethyl- 1-thia- 3,8-diaza- spiro[4.5] dec- 2-ene 35 (AF 150(S)) in paraffin oil.
  43. 103. Use of a compound according of formula (I) according to claim 1, or an enantiomer, diastereomer, racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition. WO 2003/092580 PCT/IL2003/000357 118
  44. 104. Use of the compound (S)-2-ethyl-8-methyl-1-thia- 4,8-diaza- spiro[4.5] decan- 3-one (AF267B) as a prodrug of the compound (S)-2-Ethyl-1-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292). 5 105. 2-Ethyl-4-(3-IH-indol-3-yl-propionyl)-8-methyl- 1-thia-4,8-diaza- spiro[4.5] decan-3-one in racemic form (AF704) or as the S-enantiomer thereof (AF704B), for use as a prodrug of at least one of the group of AF267B, AF292 and indole-3-propionic acid.
  45. 106. Use of a compound according to any one of claims 1 to 20, or an enantiomer, diastereomer, 10 racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition.
  46. 107. Use according to claim 106, wherein said compound is AF292 or a prodrug of AF292 or a pharmaceutically acceptable salt of either AF292 or a prodrug of AF292. 15
  47. 108. Use according to claim 107, wherein said prodrug is AF267B or a pharmaceutically acceptable salt thereof.
  48. 109. Use of a compound according to any one of claims 9 to 20, or an enantiomer, diastereomer, 20 racemate, tautomer, geometrical isomer, dimer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition for both stimulating the Ml muscarinic receptor and retarding oxidation in the vicinity of said MI muscarinic receptor.
  49. 110. Use (S)-2-Ethyl- 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza 25 spiro[4.5]decan-3-one (AF700), or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition for both stimulating the MI muscarinic receptor and activating a secretase.
  50. 111. Use of (S)-2-Ethyl- 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza 30 spiro[4.5]decan-3-one (AF700), or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition for both stimulating the Ml muscarinic receptor and antagonizing 3 secretase.
  51. 112. Use of (S)-2-Ethyl- 4-(4-fluoro-benzenesulfonyl)- 8-methyl- 1-thia- 4,8-diaza 35 spiro[4.5]decan-3-one (AF700), or an enantiomer, diastereomer, racemate, tautomer, metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition for both stimulating the MI muscarinic receptor and antagonizing y-secretase. WO 2003/092580 PCT/IL2003/000357 119
  52. 113. Use of the compound (S)-2-Ethyl-1-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292), or a metabolite or pharmaceutically acceptable salt thereof, in the preparation of a pharmaceutical composition for both stimulating the Ml muscarinic receptor and antagonizing the M3 muscarinic receptor. 5
  53. 114. Use of the compound (S)-2-ethyl-8-methyl-l-thia- 4,8-diaza- spiro[4.5] decan- 3-one (AF267B) as a prodrug of the compound (S)-2-Ethyl- I -thia-4,8-diaza-spiro[4.5] decan-3-one (AF292).
  54. 115. Use of a combination of the compounds (S)-2-ethyl- 8-methyl- 1-thia- 4,8-diaza 10 spiro[4.5]decan-3-one (AF267B) and (S)-2-Ethyl- 1-thia- 4,8-diaza- spiro[4.5] decan-3-one (AF292) in the preparation of a pharmaceutical composition for stimulating the Ml muscarinic receptor and antagonizing the M3 muscarinic receptor.
  55. 116. Use of a combination of a first compound according to claim 1 which is (S)-2-Ethyl- 1-thia 15 4,8-diaza-spiro[4.5] decan-3-one (AF292) and a second compound selected from the group consisting of compounds according to claim 1, AF267B and AF150(S), including racemates, enantiomers, diastereomers, tautomers, geometric isomers and pharmaceutically acceptable salts thereof, in the preparation of a pharmaceutical composition for stimulating the MI muscarinic receptor while minimizing adverse side-effects due to stimulation of other mAChR subtypes. 20
  56. 117. Use of 2-Ethyl-4-(3-IH-indol-3-yl-propionyl)-8-methyl- 1-thia-4,8-diaza- spiro[4.5] decan-3-one in racemic form (AF704) or as the S-enantiomer thereof (AF704B), in the preparation of a pharmaceutical composition for stimulating the Ml muscarinic receptor, retarding oxidation in the vicinity of the MI muscarinic receptor, and providing neuroprotectant activity. 25
  57. 118. Use of a compound or a mixture of compounds selected from the group consisting of compound according to claim 1, AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for inhibiting the release or synthesis of beta-amyloid peptide (AP) in a 30 mammalian cell, tissue or organism.
  58. 119. Use of a compound or a mixture of compounds selected from the group consisting of compounds according to claim 1, AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts, in the preparation of a pharmaceutical 35 composition for elevating the level of secreted form of the non-amyloidogenic amyloid precursor protein (ct-APPs) in a mammalian cell, tissue or organism.
  59. 120. Use of a compound or a mixture of compounds selected from the group consisting of compound according to claim 1, AF267B and AFI50(S), or racemates, enantiomers, geometrical isomers, WO 2003/092580 PCT/IL2003/000357 120 diasteromers, tautomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for decreasing the level of AP peptide in the brain of a mammal having an elevated level of AP3 in the brain. 5 121. Use of a compound or a mixture of compounds selected from the group consisting of compound according to claim 1, AF267 and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for inhibiting the release or synthesis of ApoE in a mammalian cell, tissue or organism. 10
  60. 122. Use of a compound or a mixture of compounds selected from the group consisting of compound according to claim 1, AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for decreasing levels of ApoE in a mammalian cell, tissue or organism. 15
  61. 123. Use of a compound or a mixture of compounds selected from the group consisting of compounds according to claim 1, AF267B and AF150(S), or racemates, enantiomers, geometrical isomers, diasteromers, tautomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for decreasing tau hyperphosphorylation in a mammalian cell, tissue or 20 organism.
  62. 124. Use of a compound or a mixture of compounds selected from the group consisting of compounds according to claim 1, AF267 and AF150(S), or racemates, enantiomers, diasteromers, tautomers, geometrical isomers and pharmaceutically acceptable salts thereof in the preparation of a 25 pharmaceutical composition for decreasing paired helical formation in a mammalian cell.
  63. 125. Use of a compound or a mixture of compounds selected from the group consisting of compounds according to claim 1, AF267 and AFI50(S), or racemates, enantiomers, diasteromers, tautomers, geometrical isomers and pharmaceutically acceptable salts thereof in the preparation of a 30 pharmaceutical composition for activating the Wnt signaling pathway in a mammalian cell, tissue or organism.
  64. 126. Use of a compound or a mixture of compounds selected from the group consisting of compounds according to claim 1, AF267 and AF150(S), or racemates, enantiomers, diasteromers, tautomers, 35 geometrical isomers and pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for inhibiting GSK301-mediated effects in a mammal.
  65. 127. Use of a compound or a mixture of compounds selected from the group consisting of compounds according to claim 1, AF267B and AF150(S), or racemates, enantiomers, diasteromers, tautomers, WO 2003/092580 PCT/IL2003/000357 121 geometrical isomers and pharmaceutically acceptable salts thereof which alone is effective as a neurotrophic agent and which acts synergistically with endogenous growth factors, i.e. neurotrophins, in the preparation of a pharmaceutical composition for enhancing the activity of neurotrophins. 5 128. Use of a combination of(a) a compound selected from the group consisting of AF267B, AF292, and AF704B, pharmaceutically acceptable salts thereof or mixtures of such compounds or salts, and (b) a compound selected from the group consisting of an immunotherapeutic compound against beta-amyloids and compounds that bind to amyloids, in the preparation of a pharmaceutical composition for treating or reducing cerebral amyloid angiopathy. 10
  66. 129. Use of AF267B, AF292, pharmaceutically acceptable salts thereof or a mixture of AF267B, AF292 or pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for the treatment of schizophrenia. 15 130. Use of AF267B, AF292, pharmaceutically acceptable salts or mixtures of AF267B, AF292 or pharmaceutically acceptable salts thereof in the preparation of a pharmaceutical composition for amelioration of symptoms of schizophrenia.
  67. 131. A compound according to claim 5 which is a pharmaceutically acceptable salt of AF504. 20
  68. 132. A compound according to claim 6 which is a pharmaceutically acceptable salt of AF292.
  69. 133. A compound according to claim 9 which is a pharmaceutically acceptable salt of AF291. 25 134. Isolated AF292 or a pharmaceutically acceptable salt thereof according to claim 6.
  70. 135. The compound AF292 or a pharmaceutically acceptable salt thereof according to claim 6, in substantially pure form. 30 136. The compound AF292 or a pharmaceutically acceptable salt thereof according to claim 6, in a purity of at least 99.9%. ee of 99.8%
  71. 137. The compound AF292 or a pharmaceutically acceptable salt thereof according to claim 6 in solid form. 35
  72. 138. The compound AF292 or a pharmaceutically acceptable salt thereof according to claim 6, whenever prepared as a solid. WO 2003/092580 PCT/IL2003/000357 122
  73. 139. The compound AF292 or a pharmaceutically acceptable salt thereof according to claim 6, whenever prepared outside a mammalian body.
  74. 140. A mixture of AF292 or a pharmaceutically acceptable salt thereof and at least one other 5 compound.
  75. 141. A mixture according to claim 140 wherein said at least one other compound is AF267B or a pharmaceutically acceptable salt thereof. 10 142. A mixture according to claim 140 wherein said at least one other compound is AF291 or a pharmaceutically acceptable salt thereof.
  76. 143. A mixture according to claim 142 wherein said mixture is a racemic mixture. 15 144. A mixture according to claim 142 wherein said mixture is not a racemic mixture.
  77. 145. A pharmaceutical composition according to claim 23, wherein said at least one compound is AF292 or a pharmaceutically acceptable salt thereof. 20 146. A pharmaceutical composition according to claim 145, further comprising at least one additional compound or pharmaceutically acceptable salt thereof.
  78. 147. A pharmaceutical composition according to claim 146, wherein said at least one additional compound or pharmaceutically acceptable salt thereof is selected from the group consisting of AF267B, 25 AF291 and pharmaceutically acceptable salts thereof.
  79. 148. A pharmaceutical composition according to claim 47, wherein said at least one compound is AF292 or a pharmaceutically acceptable salt thereof. 30 149. A pharmaceutical composition according to claim 51 comprising an Ml muscarinic receptor agonistic and M3 muscarinic receptor antagonistic efficacious amount of the compound (S)-2-Ethyl-1-thia-4,8-diaza-spiro[4.5] decan-3-one (AF292), or a pharmaceutically acceptable salt thereof, and at least one pharmaecutically acceptable carrier, diluent or excipient therefor. 35 150. A pharmaceutical composition according to claim 55, comprising an amount of a AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and at least one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of compounds of formula (I) according to claim 1, AF267B. and AFl50(S), effective to inhibit the cellular release or synthesis of AP3. I A. . - 1 t- A WO 2003/092580 PCT/IL2003/000357 123
  80. 151. A pharmaceutical composition according to claim 56, comprising an amount of AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and least one other compound or pharmaceutically acceptable salt thereof selected from the 5 group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), effective to elevate the level of the secreted form of the non-amyloidogenic amyloid precursor protein (a-APPs).
  81. 152. A pharmaceutical composition according to claim 57, comprising an amount of AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt 10 thereof and least one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), effective to decrease the level of AP3 in the brain of said mammal.
  82. 153. A pharmaceutical composition according to claim 152 wherein said elevated level of AP in the 15 brain is a result of hypercholesterolemia.
  83. 154. A pharmaceutical composition according to claim 153 wherein said elevated level of A3 in the brain is a result of cholinergic hypofunction. 20 155. A pharmaceutical composition according to claim 60, comprising an amount of AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and least one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), effective to inhibit the release or synthesis of ApoE in said mammal. 25
  84. 156. A pharmaceutical composition according to claim 155 wherein said ApoE is ApoE4.
  85. 157. A pharmaceutical composition according to claim 62, comprising an amount of AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt 30 thereof and least one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AFl50(S), effective to decrease the levels of ApoE in said mammal.
  86. 158. A pharmaceutical composition according to claim 157 wherein said ApoE is ApoE4. 35
  87. 159. A pharmaceutical composition according to claim 64 comprising AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and least one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of WO 2003/092580 124 PCT/IL2003/000357 compounds of formula (I) according to claim 1, AF267B and AF150(S), effective to inhibit tau hyperphosphorylation.
  88. 160. A pharmaceutical composition according to claim 159 wherein said tau hyperphosphorylation is 5 AP3-induced tau hyperphosphorylation.
  89. 161. A pharmaceutical composition according to claim 66 comprising AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and least one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of 10 compounds of formula (I) according to claim 1, AF267B and AF150(S), effective to inhibit tau hyperphosphorylation.
  90. 162. A pharmaceutical composition according to claim 67 comprising AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and least 15 one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), effective to inhibit Wnt abnormalities.
  91. 163. A pharmaceutical composition according to claim 68 comprising AF292 or a pharmaceutically 20 acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and least one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), effective to inhibit GSK31-mediated effects. 25 164. A pharmaceutical composition according to claim 163 wherein said GSK3-mediated effects are selected from the group consisting of tau hyperphosphorylation, apoptosis, P3-catenin degradation, and decrease in Wnt target genes.
  92. 165. A pharmaceutical composition according to claim 70 comprising AF292 or a pharmaceutically 30 acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and least one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), which alone is effective as a neurotrophic agent and which acts synergistically with said neurotrophins. 35 166. A pharmaceutical composition according to claim 71 comprising AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and least one other compound or pharmaceutically acceptable salt thereof selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF150(S), and at least one pharmaceutically acceptable carrier, diluent or excipient. WO 2003/092580 PCT/IL2003/000357 125
  93. 167. A pharmaceutical composition according to claim 76 comprising an efficacious amount of a AF292 or a pharmaceutically acceptable salt thereof, or a mixture of AF292 or a pharmaceutically acceptable salt thereof and least one other compound or pharmaceutically acceptable salt thereof 5 selected from the group consisting of compounds of formula (I) according to claim 1, AF267B and AF 150(S), and a pharmaceutically acceptable carrier, diluent or excipient.
  94. 168. A process for the preparation of AF292, comprising reacting N-protected 4-piperidone with ammonia or an ammonium salt and (S)-2-mercaptobutyric acid, and then deprotecting the ring nitrogen 10 atom.
  95. 169. The process of claim 168, further comprising reacting AF292 with an acid or base to form a pharmaceutically acceptable salt thereof. 15 170. The process of claim 168, wherein the piperidone nitrogen is protected as a carbamate.
  96. 171. The process of claim 170, wherein the N-protecting group is alkoxycarbonyl,
  97. 172. The process of claim 171, wherein the N-protecting group is tert-butoxycarbonyl. 20
  98. 173. The compound AF292 or a pharmaceutically acceptable salt thereof according to claim 6, in an enantiomeric excess of at least 99.8%.
AU2003223093A 2002-05-03 2003-05-01 Methods and compositions for treatment of central and peripheral nervous system disorders and novel compounds useful therefor Ceased AU2003223093B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US37743302P 2002-05-03 2002-05-03
US60/377,433 2002-05-03
PCT/IL2003/000357 WO2003092580A2 (en) 2002-05-03 2003-05-01 Methods and compositions for treatment of central and peripheral nervous system disorders and novel compounds useful therefor

Publications (2)

Publication Number Publication Date
AU2003223093A1 true AU2003223093A1 (en) 2003-11-17
AU2003223093B2 AU2003223093B2 (en) 2010-02-04

Family

ID=29401496

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2003223093A Ceased AU2003223093B2 (en) 2002-05-03 2003-05-01 Methods and compositions for treatment of central and peripheral nervous system disorders and novel compounds useful therefor

Country Status (15)

Country Link
US (3) US7439251B2 (en)
EP (1) EP1507765A4 (en)
JP (2) JP4584706B2 (en)
KR (2) KR101027570B1 (en)
CN (1) CN1662506A (en)
AR (1) AR039965A1 (en)
AU (1) AU2003223093B2 (en)
BR (1) BR0309875A (en)
CA (1) CA2484599A1 (en)
IL (1) IL164857A (en)
MX (1) MXPA04010921A (en)
NZ (2) NZ536610A (en)
TW (1) TWI330640B (en)
WO (1) WO2003092580A2 (en)
ZA (1) ZA200408861B (en)

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8173618B2 (en) * 2003-07-25 2012-05-08 University Of Massachusetts Formulations for reducing neuronal degeneration
CA2583342A1 (en) * 2004-10-13 2006-04-27 Merck And Co., Inc. Spiropiperidine compounds useful as beta-secretase inhibitors for the treatment of alzhermer's disease
ITRM20040606A1 (en) * 2004-12-13 2005-03-13 Sigma Tau Ind Farmaceuti USE OF ACETYL L-CARNITINE FOR THE TREATMENT OF THE FIBROMYALGY SYNDROME.
WO2007011810A1 (en) * 2005-07-18 2007-01-25 Merck & Co., Inc. Spiropiperidine beta-secretase inhibitors for the treatment of alzheimer's disease
CN101232882A (en) * 2005-07-26 2008-07-30 千寿制药株式会社 Percutaneously absorbable ophthalmic preparation
GB0607949D0 (en) * 2006-04-21 2006-05-31 Minster Res The Ltd Mono and combination therapy
ITMI20061279A1 (en) * 2006-06-30 2008-01-01 Consiglio Nazionale Ricerche SELECTIVE NICOTINIC AGONISTS FOR THE ALFA7 RECEPTOR SUBTIPLE, PROCEDURE FOR THEIR PREPARATION AND RELATED PHARMACEUTICAL COMPOSITIONS
AU2007293416A1 (en) * 2006-09-07 2008-03-13 Merck & Co., Inc. Spiropiperidine beta-secretase inhibitors for the treatment of Alzheimer's disease
EP2091328B1 (en) * 2006-10-30 2011-12-28 Merck Sharp & Dohme Corp. Spiropiperidine beta-secretase inhibitors for the treatment of alzheimer's disease
US8377954B2 (en) * 2007-01-04 2013-02-19 Merck, Sharp & Dohme, Corp Bicyclic spiropiperidine beta-secretase inhibitors for the treatment of Alzheimer's disease
US8597640B2 (en) * 2007-10-31 2013-12-03 University Of Massachusetts Lowell Over-the-counter vitamin/nutriceutical formulation that provides neuroprotection and maintains or improves cognitive performance in alzheimer's disease and normal aging
US8653068B2 (en) 2009-10-30 2014-02-18 Pain Therapeutics, Inc. Filamin A binding anti-inflammatory and analgesic
US9340558B2 (en) 2007-11-02 2016-05-17 Pain Therapeutics Inc. Filamin a binding anti-inflammatory and analgesic
US8614324B2 (en) * 2008-10-31 2013-12-24 Pain Therapeutics, Inc. Filamin A binding anti-inflammatory and analgesic
RU2476431C2 (en) 2008-01-18 2013-02-27 Эйсай Ар Энд Ди Менеджмент Ко., Лтд. Condensed aminohydrothiazine derivative
US7863291B2 (en) * 2008-04-23 2011-01-04 Bristol-Myers Squibb Company Quinuclidine compounds as alpha-7 nicotinic acetylcholine receptor ligands
US8309577B2 (en) 2008-04-23 2012-11-13 Bristol-Myers Squibb Company Quinuclidine compounds as α-7 nicotinic acetylcholine receptor ligands
AU2009277485B2 (en) * 2008-07-28 2013-05-02 Eisai R&D Management Co., Ltd. Spiroaminodihydrothiazine derivatives
JP5444240B2 (en) * 2008-07-28 2014-03-19 エーザイ・アール・アンド・ディー・マネジメント株式会社 Spiroaminodihydrothiazine derivatives
EP2332943B1 (en) * 2008-09-30 2015-04-22 Eisai R&D Management Co., Ltd. Novel fused aminodihydrothiazine derivative
US8580808B2 (en) * 2009-10-30 2013-11-12 Pain Therapeutic, Inc. Filamin A-binding anti-inflammatory analgesic
WO2010051497A1 (en) * 2008-10-31 2010-05-06 Pain Therapeutics, Inc. Filamin a binding anti-inflammatory and analgesic
BRPI1007350B8 (en) * 2009-01-26 2021-05-25 Israel Institute For Biological Res bicyclic heterocyclic spiro compounds
US9034891B2 (en) 2009-01-26 2015-05-19 Israel Institute For Biological Research Bicyclic heterocyclic spiro compounds
WO2010151609A1 (en) * 2009-06-24 2010-12-29 Photoswitch Biosciences Inc. Photoswitch-enabled ion channel assay system
GB0912778D0 (en) 2009-07-22 2009-08-26 Eisai London Res Lab Ltd Fused aminodihydro-oxazine derivatives
HUE044653T2 (en) * 2009-07-22 2019-11-28 PureTech Health LLC Compositions for treatment of disorders ameliorated by muscarinic receptor activation
US10265311B2 (en) 2009-07-22 2019-04-23 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
GB0912777D0 (en) 2009-07-22 2009-08-26 Eisai London Res Lab Ltd Fused aminodihydropyrimidone derivatives
US8278320B2 (en) 2009-10-28 2012-10-02 Bristol-Myers Squibb Company Azabicyclo[2.2.1]heptane compounds as alpha-7 nicotinic acetylcholine receptor ligands
US8507516B2 (en) 2009-10-28 2013-08-13 Bristol-Myers Squibb Company Azabicyclic compounds as alpha-7 nicotinic acetylcholine receptor ligands
US8580809B2 (en) * 2009-10-30 2013-11-12 Pain Therapeutics, Inc. Filamin A-binding anti-inflammatory analgesic
US9271986B2 (en) 2009-11-12 2016-03-01 Pharmatrophix, Inc. Crystalline forms of neurotrophin mimetic compounds and their salts
US10273219B2 (en) 2009-11-12 2019-04-30 Pharmatrophix, Inc. Crystalline forms of neurotrophin mimetic compounds and their salts
EP2556076A2 (en) * 2010-04-09 2013-02-13 Pfizer Inc. Novel sultam compounds
US8481555B2 (en) 2010-04-30 2013-07-09 Bristol-Myers Squibb Company Aza-bicyclic amine N-oxide compounds as alpha-7 nicotinic acetylcholine receptor ligand pro-drugs
GB201100181D0 (en) 2011-01-06 2011-02-23 Eisai Ltd Fused aminodihydrothiazine derivatives
GB201101139D0 (en) 2011-01-21 2011-03-09 Eisai Ltd Fused aminodihydrothiazine derivatives
GB201101140D0 (en) 2011-01-21 2011-03-09 Eisai Ltd Fused aminodihydrothiazine derivatives
JO3753B1 (en) * 2011-10-14 2021-01-31 Otsuka Pharma Co Ltd Tablet comprising 7-[4-(4-benzo[b]thiophen-4-yl-piperazin-1-yl)butoxy]-1H-quinolin-2-one or a salt thereof
TWI632921B (en) * 2011-10-19 2018-08-21 大塚製藥股份有限公司 Solution for oral administration
US20130331330A1 (en) * 2012-06-08 2013-12-12 The Board Of Trustees Of The Leland Stanford Junior University Methods of treating alzheimer's disease and other tauopathies with inhibitors of microtubule affinity regulating kinase
WO2014012054A1 (en) 2012-07-13 2014-01-16 Pain Therapeutics, Inc. Alzheimer's disease assay in a living patent
DK2882428T3 (en) 2012-07-13 2019-04-15 Pain Therapeutics Inc PROCEDURE TO INHIBIT TAU PHOSPHORIZATION
WO2014141110A2 (en) * 2013-03-14 2014-09-18 Curadev Pharma Pvt. Ltd. Aminonitriles as kynurenine pathway inhibitors
US9433604B2 (en) 2013-10-08 2016-09-06 Pain Therapeutics Inc. Method for inhibiting growth of cancer cells
CN103923105B (en) * 2014-04-17 2016-08-24 北京大学 2-indolizine Carbox amide and preparation thereof and purposes
CN105085348B (en) * 2014-05-20 2017-07-25 浙江大学 A kind of benzoic acid sulfur ester and its application
US10307409B2 (en) 2015-03-06 2019-06-04 Chase Pharmaceuticals Corporation Muscarinic combinations and their use for combating hypocholinergic disorders of the central nervous system
WO2016144727A1 (en) * 2015-03-06 2016-09-15 Chase Pharmaceuticals Corporation Peripheral-anticholinergic muscarinic agonist combination
PT3523310T (en) * 2016-10-05 2022-08-04 Nsc Therapeutics Gmbh Crystalline polymorphs of a muscarinic acetylcholine receptor agonist
US11948662B2 (en) * 2017-02-17 2024-04-02 The Regents Of The University Of California Metabolite, annotation, and gene integration system and method
US20210000807A1 (en) 2018-03-22 2021-01-07 Nsc Therapeutics Gmbh Compounds and methods for use in the treatment of microglia-mediated disorders
CA3180743A1 (en) 2018-09-28 2020-04-02 Karuna Therapeutics, Inc. Composition comprising xanomeline and trospium for treating disorders ameliorated by muscarinic receptor activation
WO2021228123A1 (en) * 2020-05-12 2021-11-18 北京广为医药科技有限公司 Compound for adjusting activity of nmda receptor, and pharmaceutical composition and use thereof
KR102245391B1 (en) 2020-10-08 2021-05-18 강현주 Bending strip apparatus and medical cable manufacturing system with the same

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE331282B (en) * 1966-12-16 1970-12-21 Yoshitomi Pharmaceutical
US3723442A (en) 1970-12-31 1973-03-27 Yoshitomi Pharmaceutical 3-oxo-1-oxa-4,8-diazaspiro(4.5)decanes
US3897425A (en) * 1974-04-01 1975-07-29 Interx Research Corp 3-Chloro-tetrahydro-1,3-oxazines or oxazolidines
JPS60184012A (en) 1984-03-02 1985-09-19 Yoshitomi Pharmaceut Ind Ltd Immuno-regulator
US4855290A (en) 1985-05-10 1989-08-08 State Of Israel, Represented By Prime Minister's Office, Israel Institute For Biological Research Derivatives of quinuclidine
US4981858A (en) 1987-08-13 1991-01-01 State Of Israel, Represented By The Prime Minister's Office, Israel Institute For Biological Research Optical isomers
US4900830A (en) 1987-10-28 1990-02-13 Israel Institute For Biological Research Process for labelling sulfur-containing compounds
US4876260A (en) 1987-10-28 1989-10-24 State Of Israel, Israel Institute Of Biological Research Oxathiolanes
US5053412A (en) 1990-04-10 1991-10-01 Israel Institute For Biological Research Spiro nitrogen-bridged heterocyclic compounds
US5852029A (en) * 1990-04-10 1998-12-22 Israel Institute For Biological Research Aza spiro compounds acting on the cholinergic system with muscarinic agonist activity
US5534520A (en) 1990-04-10 1996-07-09 Fisher; Abraham Spiro compounds containing five-membered rings
US5407938A (en) 1990-04-10 1995-04-18 Israel Institute For Biological Research Certain 1-methyl-piperidine-4-spiro-4'-(1'-3'-oxazolines) and corresponding -(1',3' thiazolines)
US6277874B1 (en) 1996-07-09 2001-08-21 University Of Cincinnati And Apologic, Incorporated Methods for the treatment of apolipoprotein E related diseases
TW453999B (en) * 1997-06-27 2001-09-11 Fujisawa Pharmaceutical Co Benzimidazole derivatives
EP1017375A2 (en) 1997-09-24 2000-07-12 Nova Molecular, Inc. Methods for increasing apoe levels for the treatment of neurodegenerative disease
GB9916562D0 (en) * 1999-07-14 1999-09-15 Pharmacia & Upjohn Spa 3-Arylsulfonyl-2-(substituted-methyl) propanoic acid derivatives as matrix metalloproteinase inhibitora
DE19962543A1 (en) * 1999-12-23 2001-07-05 Degussa Chromatographic separation of enantiomers of bicyclic lactams

Also Published As

Publication number Publication date
KR20100075700A (en) 2010-07-02
EP1507765A2 (en) 2005-02-23
KR20050025168A (en) 2005-03-11
AR039965A1 (en) 2005-03-09
NZ536610A (en) 2009-02-28
EP1507765A4 (en) 2006-11-02
CA2484599A1 (en) 2003-11-13
IL164857A (en) 2010-12-30
US7049321B2 (en) 2006-05-23
MXPA04010921A (en) 2005-05-27
IL164857A0 (en) 2005-12-18
WO2003092580A2 (en) 2003-11-13
ZA200408861B (en) 2006-07-26
US20060052406A1 (en) 2006-03-09
WO2003092580A3 (en) 2004-05-06
KR100998804B1 (en) 2010-12-06
JP2005530742A (en) 2005-10-13
AU2003223093B2 (en) 2010-02-04
JP2010184946A (en) 2010-08-26
BR0309875A (en) 2007-04-10
US20040044018A1 (en) 2004-03-04
WO2003092580B1 (en) 2004-06-24
US20080306103A1 (en) 2008-12-11
NZ560368A (en) 2008-11-28
KR101027570B1 (en) 2011-04-06
US7439251B2 (en) 2008-10-21
TWI330640B (en) 2010-09-21
CN1662506A (en) 2005-08-31
JP4759649B2 (en) 2011-08-31
TW200404808A (en) 2004-04-01
JP4584706B2 (en) 2010-11-24

Similar Documents

Publication Publication Date Title
AU2003223093B2 (en) Methods and compositions for treatment of central and peripheral nervous system disorders and novel compounds useful therefor
KR101216296B1 (en) Thienopyridinone Compounds and Methods of Treatment
US7078409B2 (en) Fused quinazoline derivatives useful as tyrosine kinase inhibitors
HUE029658T2 (en) Enantiomers of spiro-oxindole compounds and their uses as therapeutic agents
KR20150131233A (en) Substituted triazolopyridines and methods of use thereof
JP2008515907A (en) Thienopyridinone compounds and methods for their treatment
PT2356123E (en) Spiro-5,6-dihydro-4h-2,3,5,10b-tetraaza-benzo[e]azulenes
EP4339189A1 (en) Nmda receptor antagonist and use thereof
WO2018072602A1 (en) Therapeutic compounds and methods of use thereof
JP5673676B2 (en) Imidazo [1,2-a] pyridine derivatives
JP7328260B2 (en) N-substituted tetrahydrothienopyridine derivative and use thereof
TW202130643A (en) Spirocyclic o-glycoprotein-2-acetamido-2-deoxy-3-d-glucopyranosidase inhibitors
RU2776476C2 (en) N-substituted tetrahydrothienopyridine derivatives and application thereof
EA041825B1 (en) N-SUBSTITUTED TETRAHYDROTIENOPYRIDINE DERIVATIVES AND THEIR APPLICATIONS
JP5873612B2 (en) Tricyclic derivatives and their pharmaceutical uses and compositions
CN116693446A (en) Phenylurea derivative and medical application thereof

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired